1
|
Ghasemi A, Jeddi S, Kashfi K. Brain glucose metabolism: Role of nitric oxide. Biochem Pharmacol 2025; 232:116728. [PMID: 39709040 DOI: 10.1016/j.bcp.2024.116728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
One possible reason for failure in achieving optimal glycemic control in patients with type 2 diabetes (T2D) is that less attention has been paid to the brain, a fundamental player in glucose homeostasis, that consumes about 25% of total glucose utilization. In addition, animal and human studies indicate that nitric oxide (NO) is a critical player in glucose metabolism. NO synthesis from L-arginine is lower in patients with T2D, and endothelial NO synthase (eNOS)-derived NO bioavailability is lower in T2D. NO in the nervous system plays a role in neurovascular coupling (NVC) and the hypothalamic control of glucose sensing and energy homeostasis, influencing glucose utilization. This review explores NO's role in the brain's glucose metabolism. Literature indicates that glucose metabolism is different between neurons and astrocytes. Unlike neurons, astrocytes have a higher rate of glycolysis and a greater ability for lactate production. Astrocytes produce a greater amount of NO than neurons. NO inhibits mitochondrial respiration in both neurons and astrocytes and decreases intracellular ATP. NO-induced inhibition of mitochondrial respiration in neurons is not accompanied by compensatory glycolysis because phosphofructokinase 2.3 (PFK2.3), the most potent activator of PFK1 and thus glycolysis, is subjected to ubiquitylation and proteasomal degradation by cadherin-1 (Cdh1)-activated anaphase-promoting complex/cyclosome (APC/C), which leads to a low glycolytic rate in neurons. In astrocytes, NO inhibits mitochondrial respiration, but astrocytes display compensatory glycolysis by activating the adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, New York 10091, USA
| |
Collapse
|
2
|
Cao S, Guo J, Zhu D, Sun Z, Liu L, Zhang Y, Maratbek S, Wang Z, Zhang J, Li W, Ding J, Deng X, Zhang H. Brucella induced upregulation of NO promote macrophages glycolysis through the NF-κB/G6PD pathway. Int Immunopharmacol 2024; 142:113038. [PMID: 39276450 DOI: 10.1016/j.intimp.2024.113038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024]
Abstract
Increased glycolytic metabolism recently emerged as an essential process driving host defense against Brucella, but little is known about how this process is regulated during infection. We have identified a critical role for nuclear factor kappa B (NF-κB) transcription factor regulation in glycolytic switching during Brucella infection for the first time. Chromatin immunoprecipitation with next-generation sequencing for NF-κB and DNA Pull-Down revealed two novel NF-κB-binding sites in the enhancer region of the Nitric oxide (NO)production-response regulator gene glucose-6-phosphate dehydrogenase (G6PD), which is important for the switch to glycolysis during a Brucella infection. These findings demonstrate that Brucella drives metabolic reprogramming by inhibiting host oxidative phosphorylation (OXPHOS) and enhancing its glycolysis via the NF-κB-G6PD-NO-pathway. These studies provide a theoretical basis for investigating drugs or vaccines to control Brucella colonization and induction of undulant by manipulating host metabolic patterns.
Collapse
Affiliation(s)
- Shuzhu Cao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Jia Guo
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Dexin Zhu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Zhihua Sun
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Liangbo Liu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Yu Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Suleimenov Maratbek
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China; College of Veterinary, National Agricultural University of Kazakhstan, Nur Sultan, Kazakhstan
| | - Zhen Wang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Jing Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Wei Li
- Xinjiang Center for Animal Disease Prevention and Control, Urumqi, China.
| | - Jian Ding
- Xinjiang Center for Animal Disease Prevention and Control, Urumqi, China.
| | - Xingmei Deng
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| | - Hui Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China.
| |
Collapse
|
3
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
4
|
Dobolyi A, Cservenák M, Bagó AG, Chen C, Stepanova A, Paal K, Lee J, Palkovits M, Hudson G, Chinopoulos C. Cell-specific expression of key mitochondrial enzymes limits OXPHOS in astrocytes of the adult human neocortex and hippocampal formation. Commun Biol 2024; 7:1045. [PMID: 39181993 PMCID: PMC11344819 DOI: 10.1038/s42003-024-06751-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] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
The astrocyte-to-neuron lactate shuttle model entails that, upon glutamatergic neurotransmission, glycolytically derived pyruvate in astrocytes is mainly converted to lactate instead of being entirely catabolized in mitochondria. The mechanism of this metabolic rewiring and its occurrence in human brain are unclear. Here by using immunohistochemistry (4 brains) and imaging mass cytometry (8 brains) we show that astrocytes of the adult human neocortex and hippocampal formation express barely detectable amounts of mitochondrial proteins critical for performing oxidative phosphorylation (OXPHOS). These data are corroborated by queries of transcriptomes (107 brains) of neuronal versus non-neuronal cells fetched from the Allen Institute for Brain Science for genes coding for a much larger repertoire of entities contributing to OXPHOS, showing that human non-neuronal elements barely expressed mRNAs coding for such proteins. With less OXPHOS, human brain astrocytes are thus bound to produce more lactate to avoid interruption of glycolysis.
Collapse
Affiliation(s)
- Arpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eotvos Lorand University, Budapest, Hungary
| | - Melinda Cservenák
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eotvos Lorand University, Budapest, Hungary
| | - Attila G Bagó
- National Institute of Mental Health, Neurology and Neurosurgery, Department of Surgical Neurooncology, Budapest, Hungary
| | - Chun Chen
- Wellcome Centre for Mitochondrial Research, Bioscience Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Anna Stepanova
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Krisztina Paal
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Jeonghyoun Lee
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Miklós Palkovits
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
- Human Brain Tissue Bank, Semmelweis University, Budapest, Hungary
| | - Gavin Hudson
- Wellcome Centre for Mitochondrial Research, Bioscience Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Christos Chinopoulos
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
5
|
Habean ML, Kaiser KE, Williams JL. Orchestrating Stress Responses in Multiple Sclerosis: A Role for Astrocytic IFNγ Signaling. Int J Mol Sci 2024; 25:7524. [PMID: 39062765 PMCID: PMC11276796 DOI: 10.3390/ijms25147524] [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: 05/20/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease that is characterized by the infiltration of peripheral immune cells into the central nervous system (CNS), secretion of inflammatory factors, demyelination, and axonal degeneration. Inflammatory mediators such as cytokines alter cellular function and activate resident CNS cells, including astrocytes. Notably, interferon (IFN)γ is a prominent pleiotropic cytokine involved in MS that contributes to disease pathogenesis. Astrocytes are dynamic cells that respond to changes in the cellular microenvironment and are highly responsive to many cytokines, including IFNγ. Throughout the course of MS, intrinsic cell stress is initiated in response to inflammation, which can impact the pathology. It is known that cell stress is pronounced during MS; however, the specific mechanisms relating IFNγ signaling to cell stress responses in astrocytes are still under investigation. This review will highlight the current literature regarding the impact of IFNγ signaling alone and in combination with other immune mediators on astrocyte synthesis of free oxygen radicals and cell death, and cover what is understood regarding astrocytic mitochondrial dysfunction and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Maria L. Habean
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA;
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| | - Kaitlin E. Kaiser
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Doulias PT, Yang H, Andreyev AY, Dolatabadi N, Scott H, K Raspur C, Patel PR, Nakamura T, Tannenbaum SR, Ischiropoulos H, Lipton SA. S-Nitrosylation-mediated dysfunction of TCA cycle enzymes in synucleinopathy studied in postmortem human brains and hiPSC-derived neurons. Cell Chem Biol 2023; 30:965-975.e6. [PMID: 37478858 PMCID: PMC10530441 DOI: 10.1016/j.chembiol.2023.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 03/16/2023] [Accepted: 06/16/2023] [Indexed: 07/23/2023]
Abstract
A causal relationship between mitochondrial metabolic dysfunction and neurodegeneration has been implicated in synucleinopathies, including Parkinson disease (PD) and Lewy body dementia (LBD), but underlying mechanisms are not fully understood. Here, using human induced pluripotent stem cell (hiPSC)-derived neurons with mutation in the gene encoding α-synuclein (αSyn), we report the presence of aberrantly S-nitrosylated proteins, including tricarboxylic acid (TCA) cycle enzymes, resulting in activity inhibition assessed by carbon-labeled metabolic flux experiments. This inhibition principally affects α-ketoglutarate dehydrogenase/succinyl coenzyme-A synthetase, metabolizing α-ketoglutarate to succinate. Notably, human LBD brain manifests a similar pattern of aberrantly S-nitrosylated TCA enzymes, indicating the pathophysiological relevance of these results. Inhibition of mitochondrial energy metabolism in neurons is known to compromise dendritic length and synaptic integrity, eventually leading to neuronal cell death. Our evidence indicates that aberrant S-nitrosylation of TCA cycle enzymes contributes to this bioenergetic failure.
Collapse
Affiliation(s)
- Paschalis-Thomas Doulias
- Children's Hospital of Philadelphia Departments of Pediatrics and Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Chemistry and University Research Center of Ioannina, University of Ioannina, 45110 Ioannina, Greece
| | - Hongmei Yang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Changchun University of Chinese Medicine, Changchun 130021, China
| | - Alexander Y Andreyev
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nima Dolatabadi
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Henry Scott
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Charlene K Raspur
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Parth R Patel
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tomohiro Nakamura
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Steven R Tannenbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Departments of Pediatrics and Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
7
|
Liu L, Li W, Wang L, Gong P, Lyu T, Liu D, Zhang Y, Guo Y, Liu X, Tang M, Hu H, Liu C, Li B. Proteomic and metabolomic profiling of acupuncture for migraine reveals a correlative link via energy metabolism. Front Neurosci 2022; 16:1013328. [PMID: 36248663 PMCID: PMC9557737 DOI: 10.3389/fnins.2022.1013328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Migraine is a neurovascular disease with a high disability rate. Acupuncture treatment has emerged as a safe and viable alternative prophylactic therapy that can effectively alleviate the duration and frequency of migraine attacks. However, the therapeutic mechanisms underlying the effects of acupuncture are yet to be systematically elucidated. In this study, we enrolled female patients with migraine without aura (n = 20) and healthy controls (n = 10). Patients received acupuncture treatment on DU20, DU24, bilateral GB13, GB8, and GB20, applied three times per week over the course of 4 weeks for 12 sessions in total. Blood samples were collected from the median cubital vein before and after acupuncture treatment. Proteomic and metabolomic profiling was performed using liquid chromatography-mass spectrometry to determine the characteristics of differentially expressed molecules and expression of their corresponding biological pathways as well as to elucidate the pathogenesis of migraine and the biological effects underlying the treatment of migraine with acupuncture. Proteomic and metabolomic profiling of plasma samples from patients with migraine without aura before and after acupuncture treatment revealed enrichment of immune-related pathway functions and the arginine synthesis pathway. Joint pathway analyses revealed significant enrichment of the pentose phosphate and glycolysis/gluconeogenesis pathways in patients with migraine. The glycolysis/gluconeogenesis and riboflavin metabolism pathways were significantly enriched after acupuncture treatment. The expression levels of various key proteins and metabolites, including α-D-glucose, flavin adenine dinucleotide, biliverdin reductase B, and L-glutamate, were significantly differentially expressed before and after acupuncture treatment in patients with migraine without aura. Treatment of migraine with acupuncture was associated with significant changes in key molecules and pathways, indicative of physiological changes in the trigeminovascular system, glutamate neurotoxicity, and other migraine-related physiological changes. Overall, our comprehensive analysis using proteomic and metabolomic profiling demonstrates that energy metabolism may serve as a key correlative link in the occurrence of migraine and the therapeutic effects of acupuncture treatment. Our findings may facilitate the identification of diagnostic and therapeutic modalities in the ongoing search for effective treatments for migraine attacks.
Collapse
Affiliation(s)
- Lu Liu
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Weizheng Li
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Linpeng Wang
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Pengyun Gong
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Tianli Lyu
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Dapeng Liu
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yajie Zhang
- Shanxi Hospital of Integrated Traditional and Western Medicine, Taiyuan, China
| | - Yijie Guo
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Xiang Liu
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Min Tang
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Hongke Hu
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Chao Liu
- School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
- *Correspondence: Chao Liu,
| | - Bin Li
- Beijing Key Laboratory of Acupuncture Neuromodulation, Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Bin Li,
| |
Collapse
|
8
|
Busquets-García A, Bolaños JP, Marsicano G. Metabolic Messengers: endocannabinoids. Nat Metab 2022; 4:848-855. [PMID: 35817852 DOI: 10.1038/s42255-022-00600-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Arnau Busquets-García
- Cell-type mechanisms in normal and pathological behavior Research Group. IMIM-Hospital del Mar Medical Research Institute, PRBB, Barcelona, Spain.
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain.
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain.
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, Bordeaux, France.
- University of Bordeaux, Bordeaux, France.
| |
Collapse
|
9
|
Brunetta HS, Petrick HL, Momken I, Handy RM, Pignanelli C, Nunes EA, Piquereau J, Mericskay M, Holloway GP. Nitrate consumption preserves HFD-induced skeletal muscle mitochondrial ADP sensitivity and lysine acetylation: A potential role for SIRT1. Redox Biol 2022; 52:102307. [PMID: 35398714 PMCID: PMC9006675 DOI: 10.1016/j.redox.2022.102307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 03/28/2022] [Indexed: 11/26/2022] Open
Abstract
Dietary nitrate supplementation, and the subsequent serial reduction to nitric oxide, has been shown to improve glucose homeostasis in several pre-clinical models of obesity and insulin resistance. While the mechanisms remain poorly defined, the beneficial effects of nitrate appear to be partially dependent on AMPK-mediated signaling events, a central regulator of metabolism and mitochondrial bioenergetics. Since AMPK can activate SIRT1, we aimed to determine if nitrate supplementation (4 mM sodium nitrate via drinking water) improved skeletal muscle mitochondrial bioenergetics and acetylation status in mice fed a high-fat diet (HFD: 60% fat). Consumption of HFD induced whole-body glucose intolerance, and within muscle attenuated insulin-induced Akt phosphorylation, mitochondrial ADP sensitivity (higher apparent Km), submaximal ADP-supported respiration, mitochondrial hydrogen peroxide (mtH2O2) production in the presence of ADP and increased cellular protein carbonylation alongside mitochondrial-specific acetylation. Consumption of nitrate partially preserved glucose tolerance and, within skeletal muscle, normalized insulin-induced Akt phosphorylation, mitochondrial ADP sensitivity, mtH2O2, protein carbonylation and global mitochondrial acetylation status. Nitrate also prevented the HFD-mediated reduction in SIRT1 protein, and interestingly, the positive effects of nitrate ingestion on glucose homeostasis and mitochondrial acetylation levels were abolished in SIRT1 inducible knock-out mice, suggesting SIRT1 is required for the beneficial effects of dietary nitrate. Altogether, dietary nitrate preserves mitochondrial ADP sensitivity and global lysine acetylation in HFD-fed mice, while in the absence of SIRT1, the effects of nitrate on glucose tolerance and mitochondrial acetylation were abrogated.
Collapse
|
10
|
Harguindey S, Alfarouk K, Polo Orozco J, Reshkin SJ, Devesa J. Hydrogen Ion Dynamics as the Fundamental Link between Neurodegenerative Diseases and Cancer: Its Application to the Therapeutics of Neurodegenerative Diseases with Special Emphasis on Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23052454. [PMID: 35269597 PMCID: PMC8910484 DOI: 10.3390/ijms23052454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
The pH-related metabolic paradigm has rapidly grown in cancer research and treatment. In this contribution, this recent oncological perspective has been laterally assessed for the first time in order to integrate neurodegeneration within the energetics of the cancer acid-base conceptual frame. At all levels of study (molecular, biochemical, metabolic, and clinical), the intimate nature of both processes appears to consist of opposite mechanisms occurring at the far ends of a physiopathological intracellular pH/extracellular pH (pHi/pHe) spectrum. This wide-ranging original approach now permits an increase in our understanding of these opposite processes, cancer and neurodegeneration, and, as a consequence, allows us to propose new avenues of treatment based upon the intracellular and microenvironmental hydrogen ion dynamics regulating and deregulating the biochemistry and metabolism of both cancer and neural cells. Under the same perspective, the etiopathogenesis and special characteristics of multiple sclerosis (MS) is an excellent model for the study of neurodegenerative diseases and, utilizing this pioneering approach, we find that MS appears to be a metabolic disease even before an autoimmune one. Furthermore, within this paradigm, several important aspects of MS, from mitochondrial failure to microbiota functional abnormalities, are analyzed in depth. Finally, and for the first time, a new and integrated model of treatment for MS can now be advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Division of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
- Correspondence: ; Tel.: +34-629-047-141
| | - Khalid Alfarouk
- Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan;
| | - Julián Polo Orozco
- Division of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
11
|
Beard E, Lengacher S, Dias S, Magistretti PJ, Finsterwald C. Astrocytes as Key Regulators of Brain Energy Metabolism: New Therapeutic Perspectives. Front Physiol 2022; 12:825816. [PMID: 35087428 PMCID: PMC8787066 DOI: 10.3389/fphys.2021.825816] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes play key roles in the regulation of brain energy metabolism, which has a major impact on brain functions, including memory, neuroprotection, resistance to oxidative stress and homeostatic tone. Energy demands of the brain are very large, as they continuously account for 20–25% of the whole body’s energy consumption. Energy supply of the brain is tightly linked to neuronal activity, providing the origin of the signals detected by the widely used functional brain imaging techniques such as functional magnetic resonance imaging and positron emission tomography. In particular, neuroenergetic coupling is regulated by astrocytes through glutamate uptake that triggers astrocytic aerobic glycolysis and leads to glucose uptake and lactate release, a mechanism known as the Astrocyte Neuron Lactate Shuttle. Other neurotransmitters such as noradrenaline and Vasoactive Intestinal Peptide mobilize glycogen, the reserve for glucose exclusively localized in astrocytes, also resulting in lactate release. Lactate is then transferred to neurons where it is used, after conversion to pyruvate, as a rapid energy substrate, and also as a signal that modulates neuronal excitability, homeostasis, and the expression of survival and plasticity genes. Importantly, glycolysis in astrocytes and more generally cerebral glucose metabolism progressively deteriorate in aging and age-associated neurodegenerative diseases such as Alzheimer’s disease. This decreased glycolysis actually represents a common feature of several neurological pathologies. Here, we review the critical role of astrocytes in the regulation of brain energy metabolism, and how dysregulation of astrocyte-mediated metabolic pathways is involved in brain hypometabolism. Further, we summarize recent efforts at preclinical and clinical stages to target brain hypometabolism for the development of new therapeutic interventions in age-related neurodegenerative diseases.
Collapse
|
12
|
Wevers NR, Nair AL, Fowke TM, Pontier M, Kasi DG, Spijkers XM, Hallard C, Rabussier G, van Vught R, Vulto P, de Vries HE, Lanz HL. Modeling ischemic stroke in a triculture neurovascular unit on-a-chip. Fluids Barriers CNS 2021; 18:59. [PMID: 34906183 PMCID: PMC8670153 DOI: 10.1186/s12987-021-00294-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/30/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND In ischemic stroke, the function of the cerebral vasculature is impaired. This vascular structure is formed by the so-called neurovascular unit (NVU). A better understanding of the mechanisms involved in NVU dysfunction and recovery may lead to new insights for the development of highly sought therapeutic approaches. To date, there remains an unmet need for complex human in vitro models of the NVU to study ischemic events seen in the human brain. METHODS We here describe the development of a human NVU on-a-chip model using a platform that allows culture of 40 chips in parallel. The model comprises a perfused vessel of primary human brain endothelial cells in co-culture with induced pluripotent stem cell derived astrocytes and neurons. Ischemic stroke was mimicked using a threefold approach that combines chemical hypoxia, hypoglycemia, and halted perfusion. RESULTS Immunofluorescent staining confirmed expression of endothelial adherens and tight junction proteins, as well as astrocytic and neuronal markers. In addition, the model expresses relevant brain endothelial transporters and shows spontaneous neuronal firing. The NVU on-a-chip model demonstrates tight barrier function, evidenced by retention of small molecule sodium fluorescein in its lumen. Exposure to the toxic compound staurosporine disrupted the endothelial barrier, causing reduced transepithelial electrical resistance and increased permeability to sodium fluorescein. Under stroke mimicking conditions, brain endothelial cells showed strongly reduced barrier function (35-fold higher apparent permeability) and 7.3-fold decreased mitochondrial potential. Furthermore, levels of adenosine triphosphate were significantly reduced on both the blood- and the brain side of the model (4.8-fold and 11.7-fold reduction, respectively). CONCLUSIONS The NVU on-a-chip model presented here can be used for fundamental studies of NVU function in stroke and other neurological diseases and for investigation of potential restorative therapies to fight neurological disorders. Due to the platform's relatively high throughput and compatibility with automation, the model holds potential for drug compound screening.
Collapse
Affiliation(s)
- Nienke R Wevers
- MIMETAS BV, Leiden, The Netherlands. .,Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Arya Lekshmi Nair
- MIMETAS BV, Leiden, The Netherlands.,Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | | | | | | | - Xandor M Spijkers
- MIMETAS BV, Leiden, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | | | - Gwenaëlle Rabussier
- MIMETAS BV, Leiden, The Netherlands.,CARIM School of Cardiovascular Diseases, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | | | | | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | | |
Collapse
|
13
|
Bonvento G, Bolaños JP. Astrocyte-neuron metabolic cooperation shapes brain activity. Cell Metab 2021; 33:1546-1564. [PMID: 34348099 DOI: 10.1016/j.cmet.2021.07.006] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/11/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022]
Abstract
The brain has almost no energy reserve, but its activity coordinates organismal function, a burden that requires precise coupling between neurotransmission and energy metabolism. Deciphering how the brain accomplishes this complex task is crucial to understand central facets of human physiology and disease mechanisms. Each type of neural cell displays a peculiar metabolic signature, forcing the intercellular exchange of metabolites that serve as both energy precursors and paracrine signals. The paradigm of this biological feature is the astrocyte-neuron couple, in which the glycolytic metabolism of astrocytes contrasts with the mitochondrial oxidative activity of neurons. Astrocytes generate abundant mitochondrial reactive oxygen species and shuttle to neurons glycolytically derived metabolites, such as L-lactate and L-serine, which sustain energy needs, conserve redox status, and modulate neurotransmitter-receptor activity. Conversely, early disruption of this metabolic cooperation may contribute to the initiation or progression of several neurological diseases, thus requiring innovative therapies to preserve brain energetics.
Collapse
Affiliation(s)
- Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Institute of Biomedical Research of Salamanca, Salamanca, Spain
| |
Collapse
|
14
|
Parodi-Rullán R, Sone JY, Fossati S. Endothelial Mitochondrial Dysfunction in Cerebral Amyloid Angiopathy and Alzheimer's Disease. J Alzheimers Dis 2020; 72:1019-1039. [PMID: 31306129 DOI: 10.3233/jad-190357] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Cerebrovascular dysfunction is one of the earliest events in the pathogenesis of AD, as well as in vascular and mixed dementias. Cerebral amyloid angiopathy (CAA), the deposition of amyloid around cerebral vessels, is observed in up to 90% of AD patients and in approximately 50% of elderly individuals over 80 years of age. CAA is a strong contributor to vascular dysfunction in AD. CAA-laden brain vessels are characterized by dysfunctional hemodynamics and leaky blood-brain barrier (BBB), contributing to clearance failure and further accumulation of amyloid-β (Aβ) in the cerebrovasculature and brain parenchyma. Mitochondrial dysfunction is increasingly recognized as an important early initiator of the pathogenesis of AD and CAA. The objective of this review is to discuss the effects of Aβ on cerebral microvascular cell function, focusing on its impact on endothelial mitochondria. After introducing CAA and its etiology and genetic risk factors, we describe the pathological relationship between cerebrovascular amyloidosis and brain microvascular endothelial cell dysfunction, critically analyzing its roles in disease progression, hypoperfusion, and BBB integrity. Then, we focus on discussing the effect of Aβ challenge on endothelial mitochondrial dysfunction pathways, and their contribution to the progression of neurovascular dysfunction in AD and dementia. Finally, we report potential pharmacological and non-pharmacological mitochondria-targeted therapeutic strategies which may help prevent or delay cerebrovascular failure.
Collapse
Affiliation(s)
- Rebecca Parodi-Rullán
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Je Yeong Sone
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, NY, USA
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
15
|
Palmieri EM, McGinity C, Wink DA, McVicar DW. Nitric Oxide in Macrophage Immunometabolism: Hiding in Plain Sight. Metabolites 2020; 10:metabo10110429. [PMID: 33114647 PMCID: PMC7693038 DOI: 10.3390/metabo10110429] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric Oxide (NO) is a soluble endogenous gas with various biological functions like signaling, and working as an effector molecule or metabolic regulator. In response to inflammatory signals, immune myeloid cells, like macrophages, increase production of cytokines and NO, which is important for pathogen killing. Under these proinflammatory circumstances, called “M1”, macrophages undergo a series of metabolic changes including rewiring of their tricarboxylic acid (TCA) cycle. Here, we review findings indicating that NO, through its interaction with heme and non-heme metal containing proteins, together with components of the electron transport chain, functions not only as a regulator of cell respiration, but also a modulator of intracellular cell metabolism. Moreover, diverse effects of NO and NO-derived reactive nitrogen species (RNS) involve precise interactions with different targets depending on concentration, temporal, and spatial restrictions. Although the role of NO in macrophage reprogramming has been in evidence for some time, current models have largely minimized its importance. It has, therefore, been hiding in plain sight. A review of the chemical properties of NO, past biochemical studies, and recent publications, necessitates that mechanisms of macrophage TCA reprogramming during stimulation must be re-imagined and re-interpreted as mechanistic results of NO exposure. The revised model of metabolic rewiring we describe here incorporates many early findings regarding NO biochemistry and brings NO out of hiding and to the forefront of macrophages immunometabolism.
Collapse
|
16
|
Brunetta HS, Politis-Barber V, Petrick HL, Dennis KMJH, Kirsh AJ, Barbeau PA, Nunes EA, Holloway GP. Nitrate attenuates high fat diet-induced glucose intolerance in association with reduced epididymal adipose tissue inflammation and mitochondrial reactive oxygen species emission. J Physiol 2020; 598:3357-3371. [PMID: 32449521 DOI: 10.1113/jp279455] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/19/2020] [Indexed: 12/20/2023] Open
Abstract
KEY POINTS Dietary nitrate is a prominent therapeutic strategy to mitigate some metabolic deleterious effects related to obesity. Mitochondrial dysfunction is causally linked to adipose tissue inflammation and insulin resistance. Whole-body glucose tolerance is prevented by nitrate independent of body weight and energy expenditure. Dietary nitrate reduces epididymal adipose tissue inflammation and mitochondrial reactive oxygen species emission while preserving insulin signalling. Metabolic beneficial effects of nitrate consumption are associated with improvements in mitochondrial redox balance in hypertrophic adipose tissue. ABSTRACT Evidence has accumulated to indicate that dietary nitrate alters energy expenditure and the metabolic derangements associated with a high fat diet (HFD), but the mechanism(s) of action remain incompletely elucidated. Therefore, we aimed to determine if dietary nitrate (4 mm sodium nitrate via drinking water) could prevent HFD-mediated glucose intolerance in association with improved mitochondrial bioenergetics within both white (WAT) and brown (BAT) adipose tissue in mice. HFD feeding caused glucose intolerance (P < 0.05) and increased body weight. As a result of higher body weight, energy expenditure increased proportionally. HFD-fed mice displayed greater mitochondrial uncoupling and a twofold increase in uncoupling protein 1 content within BAT. Within epididymal white adipose tissue (eWAT), HFD increased cell size (i.e. hypertrophy), mitochondrial H2 O2 emission, oxidative stress, c-Jun N-terminal kinase phosphorylation and leucocyte infiltration, and induced insulin resistance. Remarkably, dietary nitrate consumption attenuated and/or mitigated all these responses, including rendering mitochondria more coupled within BAT, and normalizing mitochondrial H2 O2 emission and insulin-mediated Akt-Thr308 phosphorylation within eWAT. Intriguingly, the positive effects of dietary nitrate appear to be independent of eWAT mitochondrial respiratory capacity and content. Altogether, these data suggest that dietary nitrate attenuates the development of HFD-induced insulin resistance in association with attenuating WAT inflammation and redox balance, independent of changes in either WAT or BAT mitochondrial respiratory capacity/content.
Collapse
Affiliation(s)
- Henver S Brunetta
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Valerie Politis-Barber
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Heather L Petrick
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kaitlyn M J H Dennis
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Aleah J Kirsh
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Pierre-Andre Barbeau
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Everson A Nunes
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
17
|
Hill RL, Singh IN, Wang JA, Kulbe JR, Hall ED. Protective effects of phenelzine administration on synaptic and non-synaptic cortical mitochondrial function and lipid peroxidation-mediated oxidative damage following TBI in young adult male rats. Exp Neurol 2020; 330:113322. [PMID: 32325157 DOI: 10.1016/j.expneurol.2020.113322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) results in mitochondrial dysfunction and induction of lipid peroxidation (LP). Lipid peroxidation-derived neurotoxic aldehydes such as 4-HNE and acrolein bind to mitochondrial proteins, inducing additional oxidative damage and further exacerbating mitochondrial dysfunction and LP. Mitochondria are heterogeneous, consisting of both synaptic and non-synaptic populations, with synaptic mitochondria being more vulnerable to injury-dependent consequences. The goal of these studies was to explore the hypothesis that interrupting secondary oxidative damage following TBI using phenelzine (PZ), an aldehyde scavenger, would preferentially protect synaptic mitochondria against LP-mediated damage in a dose- and time-dependent manner. Male Sprague-Dawley rats received a severe (2.2 mm) controlled cortical impact (CCI)-TBI. PZ (3-30 mg/kg) was administered subcutaneously (subQ) at different times post-injury. We found PZ treatment preserves both synaptic and non-synaptic mitochondrial bioenergetics at 24 h and that this protection is partially maintained out to 72 h post-injury using various dosing regimens. The results from these studies indicate that the therapeutic window for the first dose of PZ is likely within the first hour after injury, and the window for administration of the second dose seems to fall between 12 and 24 h. Administration of PZ was able to significantly improve mitochondrial respiration compared to vehicle-treated animals across various states of respiration for both the non-synaptic and synaptic mitochondria. The synaptic mitochondria appear to respond more robustly to PZ treatment than the non-synaptic, and further experimentation will need to be done to further understand these effects in the context of TBI.
Collapse
Affiliation(s)
- Rachel L Hill
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America.
| | - Indrapal N Singh
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America; Department of Neuroscience, 741 S. Limestone St, Lexington, KY 40536-0509, United States of America
| | - Juan A Wang
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America
| | - Jacqueline R Kulbe
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America
| | - Edward D Hall
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America; Department of Neuroscience, 741 S. Limestone St, Lexington, KY 40536-0509, United States of America
| |
Collapse
|
18
|
Laranjinha J, Nunes C, Ledo A, Lourenço C, Rocha B, Barbosa RM. The Peculiar Facets of Nitric Oxide as a Cellular Messenger: From Disease-Associated Signaling to the Regulation of Brain Bioenergetics and Neurovascular Coupling. Neurochem Res 2020; 46:64-76. [PMID: 32193753 DOI: 10.1007/s11064-020-03015-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
In this review, we address the regulatory and toxic role of ·NO along several pathways, from the gut to the brain. Initially, we address the role on ·NO in the regulation of mitochondrial respiration with emphasis on the possible contribution to Parkinson's disease via mechanisms that involve its interaction with a major dopamine metabolite, DOPAC. In parallel with initial discoveries of the inhibition of mitochondrial respiration by ·NO, it became clear the potential for toxic ·NO-mediated mechanisms involving the production of more reactive species and the post-translational modification of mitochondrial proteins. Accordingly, we have proposed a novel mechanism potentially leading to dopaminergic cell death, providing evidence that NO synergistically interact with DOPAC in promoting cell death via mechanisms that involve GSH depletion. The modulatory role of NO will be then briefly discussed as a master regulator on brain energy metabolism. The energy metabolism in the brain is central to the understanding of brain function and disease. The core role of ·NO in the regulation of brain metabolism and vascular responses is further substantiated by discussing its role as a mediator of neurovascular coupling, the increase in local microvessels blood flow in response to spatially restricted increase of neuronal activity. The many facets of NO as intracellular and intercellular messenger, conveying information associated with its spatial and temporal concentration dynamics, involve not only the discussion of its reactions and potential targets on a defined biological environment but also the regulation of its synthesis by the family of nitric oxide synthases. More recently, a novel pathway, out of control of NOS, has been the subject of a great deal of controversy, the nitrate:nitrite:NO pathway, adding new perspectives to ·NO biology. Thus, finally, this novel pathway will be addressed in connection with nitrate consumption in the diet and the beneficial effects of protein nitration by reactive nitrogen species.
Collapse
Affiliation(s)
- João Laranjinha
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal. .,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal.
| | - Carla Nunes
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Ana Ledo
- Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Cátia Lourenço
- Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Bárbara Rocha
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| | - Rui M Barbosa
- Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Pólo 1, 3000-504, Coimbra, Portugal
| |
Collapse
|
19
|
Palmieri EM, Gonzalez-Cotto M, Baseler WA, Davies LC, Ghesquière B, Maio N, Rice CM, Rouault TA, Cassel T, Higashi RM, Lane AN, Fan TWM, Wink DA, McVicar DW. Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nat Commun 2020; 11:698. [PMID: 32019928 PMCID: PMC7000728 DOI: 10.1038/s41467-020-14433-7] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/16/2019] [Indexed: 01/24/2023] Open
Abstract
Profound metabolic changes are characteristic of macrophages during classical activation and have been implicated in this phenotype. Here we demonstrate that nitric oxide (NO) produced by murine macrophages is responsible for TCA cycle alterations and citrate accumulation associated with polarization. 13C tracing and mitochondrial respiration experiments map NO-mediated suppression of metabolism to mitochondrial aconitase (ACO2). Moreover, we find that inflammatory macrophages reroute pyruvate away from pyruvate dehydrogenase (PDH) in an NO-dependent and hypoxia-inducible factor 1α (Hif1α)-independent manner, thereby promoting glutamine-based anaplerosis. Ultimately, NO accumulation leads to suppression and loss of mitochondrial electron transport chain (ETC) complexes. Our data reveal that macrophages metabolic rewiring, in vitro and in vivo, is dependent on NO targeting specific pathways, resulting in reduced production of inflammatory mediators. Our findings require modification to current models of macrophage biology and demonstrate that reprogramming of metabolism should be considered a result rather than a mediator of inflammatory polarization. Production of inflammatory mediators by M1-polarized macrophages is thought to rely on suppression of mitochondrial metabolism in favor of glycolysis. Refining this concept, here the authors define metabolic targets of nitric oxide as responsible for the mitochondrial rewiring resulting from polarization.
Collapse
Affiliation(s)
- Erika M Palmieri
- Leukocyte Signaling Section, Cancer & Inflammation Program, National Cancer Institute, Frederick, MD, USA
| | - Marieli Gonzalez-Cotto
- Leukocyte Signaling Section, Cancer & Inflammation Program, National Cancer Institute, Frederick, MD, USA
| | - Walter A Baseler
- Leukocyte Signaling Section, Cancer & Inflammation Program, National Cancer Institute, Frederick, MD, USA
| | - Luke C Davies
- Leukocyte Signaling Section, Cancer & Inflammation Program, National Cancer Institute, Frederick, MD, USA.,Division of Infection & Immunity, School of Medicine, Cardiff University, Tenovus Building, Heath Park, Cardiff, CF14 4XN, UK
| | - Bart Ghesquière
- Metabolomics Expertise Center, Vesalius Research Center, VIB, 3000, Leuven, Belgium.,Metabolomics Expertise Center, Department of Oncology, KU Leuven, 3000, Leuven, Belgium
| | - Nunziata Maio
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Christopher M Rice
- Leukocyte Signaling Section, Cancer & Inflammation Program, National Cancer Institute, Frederick, MD, USA.,School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Tracey A Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Teresa Cassel
- Department of Toxicology and Cancer Biology and Markey Cancer Center and Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Richard M Higashi
- Department of Toxicology and Cancer Biology and Markey Cancer Center and Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Andrew N Lane
- Department of Toxicology and Cancer Biology and Markey Cancer Center and Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Teresa W-M Fan
- Department of Toxicology and Cancer Biology and Markey Cancer Center and Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, USA
| | - David A Wink
- Chemical and Molecular Inflammation Section, Cancer and Inflammation Program, National Cancer Institute, Frederick, MD, USA
| | - Daniel W McVicar
- Leukocyte Signaling Section, Cancer & Inflammation Program, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
20
|
Ng X, Sadeghian M, Heales S, Hargreaves IP. Assessment of Mitochondrial Dysfunction in Experimental Autoimmune Encephalomyelitis (EAE) Models of Multiple Sclerosis. Int J Mol Sci 2019; 20:ijms20204975. [PMID: 31600882 PMCID: PMC6829485 DOI: 10.3390/ijms20204975] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) that involves the autoreactive T-cell attack on axonal myelin sheath. Lesions or plaques formed as a result of repeated damage and repair mechanisms lead to impaired relay of electrical impulses along the nerve, manifesting as clinical symptoms of MS. Evidence from studies in experimental autoimmune encephalomyelitis (EAE) models of MS strongly suggests that mitochondrial dysfunction presents at the onset of disease and throughout the disease course. The aim of this study was to determine if mitochondrial dysfunction occurs before clinical symptoms arise, and whether this is confined to the CNS. EAE was induced in C57B/L6 mice, and citrate synthase and mitochondrial respiratory chain (MRC) complex I–IV activities were assayed at presymptomatic (3 or 10 days post first immunisation (3 or 10 DPI)) and asymptomatic (17 days post first immunisation (17 DPI) time-points in central nervous system (CNS; spinal cord) and peripheral (liver and jaw muscle) tissues. Samples from animals immunised with myelin oligodendrocyte glycoprotein (MOG) as EAE models were compared with control animals immunised with adjuvant (ADJ) only. Significant changes in MOG compared to control ADJ animals in MRC complex I activity occurred only at presymptomatic stages, with an increase in the spinal cord at 10 DPI (87.9%), an increase at 3 DPI (25.6%) and decrease at 10 DPI (22.3%) in the jaw muscle, and an increase in the liver at 10 DPI (71.5%). MRC complex II/III activity changes occurred at presymptomatic and the asymptomatic stages of the disease, with a decrease occurring in the spinal cord at 3 DPI (87.6%) and an increase at 17 DPI (36.7%), increase in the jaw muscle at 10 DPI (25.4%), and an increase at 3 DPI (75.2%) and decrease at 17 DPI (95.7%) in the liver. Citrate synthase activity was also significantly decreased at 10 DPI (27.3%) in the liver. No significant changes were observed in complex IV across all three tissues assayed. Our findings reveal evidence that mitochondrial dysfunction is present at the asymptomatic stages in the EAE model of MS, and that the changes in MRC enzyme activities are tissue-specific and are not confined to the CNS.
Collapse
Affiliation(s)
- Xiulin Ng
- UCL Institute of Neurology, Queen Square, University College London, London WC1N 3BG, UK.
- University Medical Center, 79106 Freiburg im Breisgau, Germany.
| | - Mona Sadeghian
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London WC1N 3BG, UK.
- Global Clinical Development, Actelion, High Wycombe HP12 4DP, UK.
| | - Simon Heales
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
- Neurometabolic Unit, National Hospital, London WC1N 3BG, UK.
| | - Iain P Hargreaves
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
- Neurometabolic Unit, National Hospital, London WC1N 3BG, UK.
- School of Pharmacy and Biomolecular Sciences, Byrom Street, Liverpool John Moores University, Liverpool L3 3AF, UK.
| |
Collapse
|
21
|
The metabolic face of migraine - from pathophysiology to treatment. Nat Rev Neurol 2019; 15:627-643. [PMID: 31586135 DOI: 10.1038/s41582-019-0255-4] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
Migraine can be regarded as a conserved, adaptive response that occurs in genetically predisposed individuals with a mismatch between the brain's energy reserve and workload. Given the high prevalence of migraine, genotypes associated with the condition seem likely to have conferred an evolutionary advantage. Technological advances have enabled the examination of different aspects of cerebral metabolism in patients with migraine, and complementary animal research has highlighted possible metabolic mechanisms in migraine pathophysiology. An increasing amount of evidence - much of it clinical - suggests that migraine is a response to cerebral energy deficiency or oxidative stress levels that exceed antioxidant capacity and that the attack itself helps to restore brain energy homeostasis and reduces harmful oxidative stress levels. Greater understanding of metabolism in migraine offers novel therapeutic opportunities. In this Review, we describe the evidence for abnormalities in energy metabolism and mitochondrial function in migraine, with a focus on clinical data (including neuroimaging, biochemical, genetic and therapeutic studies), and consider the relationship of these abnormalities with the abnormal sensory processing and cerebral hyper-responsivity observed in migraine. We discuss experimental data to consider potential mechanisms by which metabolic abnormalities could generate attacks. Finally, we highlight potential treatments that target cerebral metabolism, such as nutraceuticals, ketone bodies and dietary interventions.
Collapse
|
22
|
Burmistrova O, Olias-Arjona A, Lapresa R, Jimenez-Blasco D, Eremeeva T, Shishov D, Romanov S, Zakurdaeva K, Almeida A, Fedichev PO, Bolaños JP. Targeting PFKFB3 alleviates cerebral ischemia-reperfusion injury in mice. Sci Rep 2019; 9:11670. [PMID: 31406177 PMCID: PMC6691133 DOI: 10.1038/s41598-019-48196-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
The glycolytic rate in neurons is low in order to allow glucose to be metabolized through the pentose-phosphate pathway (PPP), which regenerates NADPH to preserve the glutathione redox status and survival. This is controlled by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3), the pro-glycolytic enzyme that forms fructose-2,6-bisphosphate, a powerful allosteric activator of 6-phosphofructo-1-kinase. In neurons, PFKFB3 protein is physiologically inactive due to its proteasomal degradation. However, upon an excitotoxic stimuli, PFKFB3 becomes stabilized to activate glycolysis, thus hampering PPP mediated protection of redox status leading to neurodegeneration. Here, we show that selective inhibition of PFKFB3 activity by the small molecule AZ67 prevents the NADPH oxidation, redox stress and apoptotic cell death caused by the activation of glycolysis triggered upon excitotoxic and oxygen-glucose deprivation/reoxygenation models in mouse primary neurons. Furthermore, in vivo administration of AZ67 to mice significantly alleviated the motor discoordination and brain infarct injury in the middle carotid artery occlusion ischemia/reperfusion model. These results show that pharmacological inhibition of PFKFB3 is a suitable neuroprotective therapeutic strategy in excitotoxic-related disorders such as stroke.
Collapse
Affiliation(s)
| | - Ana Olias-Arjona
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
| | - Rebeca Lapresa
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Daniel Jimenez-Blasco
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | | | | | | | | | - Angeles Almeida
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | | | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.
| |
Collapse
|
23
|
Tang X, Li Z, Zhang W, Yao Z. Nitric oxide might be an inducing factor in cognitive impairment in Alzheimer's disease via downregulating the monocarboxylate transporter 1. Nitric Oxide 2019; 91:35-41. [PMID: 31326499 DOI: 10.1016/j.niox.2019.07.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/08/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a typical neurodegenerative disease in central nervous system (CNS). Generally speaking, patients with severe AD are often accompanied with cognitive impairment. Oligodendrocytes (OLs) are myelin-forming cells in CNS, and myelin injury potentially has something to do with the cognitive impairment in AD. Based on the previous experimental studies, it has been recognized that nitric oxide (NO), as a signaling molecule, might have an influence on the axon and myelin by affecting the energy transport mechanism of OLs through monocarboxylate transporter 1 (MCT1). Interestingly, a novel model of cell signaling----axo-myelinic synapse (AMS) has been put forward. In the context of this model, chances are that a new way is established in which NO can influence the pathogenesis of AD by down-regulating the expression of MCT1. As a consequence, it may provide attractive prospective and underlying drug targeting effects for the treatment of AD.
Collapse
Affiliation(s)
- Xiaoyi Tang
- Department of Physiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Luliang Military Airport Hospital, Yunnan, 655699, China
| | - Zhuang Li
- Luliang Military Airport Hospital, Yunnan, 655699, China
| | - Weiwei Zhang
- Department of Physiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Zhongxiang Yao
- Department of Physiology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
24
|
Thiele RH, Osuru HP, Paila U, Ikeda K, Zuo Z. Impact of inflammation on brain subcellular energetics in anesthetized rats. BMC Neurosci 2019; 20:34. [PMID: 31307382 PMCID: PMC6631861 DOI: 10.1186/s12868-019-0514-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Emerging data suggests that volatile anesthetic agents may have organ protection properties in the setting of critical illness. The purpose of this study was to better understand the effect of inflammation on cerebral subcellular energetics in animals exposed to two different anesthetic agents—a GABA agonist (propofol) and a volatile agent (isoflurane). Results Forty-eight Sprague–Dawley rats were anesthetized with isoflurane or propofol. In each group, rats were randomized to celiotomy and closure (sham) or cecal ligation and puncture (inflammation [sepsis model]) for 8 h. Brain tissue oxygen saturation and the oxidation state of cytochrome aa3 were measured. Brain tissue was extracted using the freeze-blow technique. All rats experienced progressive increases in tissue oxygenation and cytochrome aa3 reduction over time. Inflammation had no impact on cytochrome aa3, but isoflurane caused significant cytochrome aa3 reduction. During isoflurane (not propofol) anesthesia, inflammation led to an increase in lactate (+ 0.64 vs. − 0.80 mEq/L, p = 0.0061). There were no differences in ADP:ATP ratios between groups. In the isoflurane (not propofol) group, inflammation increased the expression of hypoxia-inducible factor-1α (62%, p = 0.0012), heme oxygenase-1 (67%, p = 0.0011), and inducible nitric oxide synthase (31%, p = 0.023) in the brain. Animals exposed to inflammation and isoflurane (but not propofol) exhibited increased expression of protein carbonyls (9.2 vs. 7.0 nM/mg protein, p = 0.0050) and S-nitrosylation (49%, p = 0.045) in the brain. RNA sequencing identified an increase in heat shock protein 90 and NF-κβ inhibitor mRNA in the inflammation/isoflurane group. Conclusions In the setting of inflammation, rats exposed to isoflurane show increased hypoxia-inducible factor-1α expression despite a lack of hypoxia, increased oxidative stress in the brain, and increased serum lactate, all of which suggest a relative increase in anaerobic metabolism compared to propofol. Differences in oxidative stress as well as heat shock protein 90 and NF-κβ inhibitor may account for the differential expression of cerebral hypoxia-inducible factor-1α during inflammation. Electronic supplementary material The online version of this article (10.1186/s12868-019-0514-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert H Thiele
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710, Charlottesville, VA, 22908-0710, USA.
| | - Hari P Osuru
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710, Charlottesville, VA, 22908-0710, USA
| | - Umadevi Paila
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, USA
| | - Keita Ikeda
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710, Charlottesville, VA, 22908-0710, USA
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710, Charlottesville, VA, 22908-0710, USA
| |
Collapse
|
25
|
Carter SF, Herholz K, Rosa-Neto P, Pellerin L, Nordberg A, Zimmer ER. Astrocyte Biomarkers in Alzheimer's Disease. Trends Mol Med 2019; 25:77-95. [PMID: 30611668 DOI: 10.1016/j.molmed.2018.11.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023]
Abstract
Astrocytic contributions to Alzheimer's disease (AD) progression were, until recently, largely overlooked. Astrocytes are integral to normal brain function and astrocyte reactivity is an early feature of AD, potentially providing a promising target for preclinical diagnosis and treatment. Several in vivo AD biomarkers already exist, but presently there is a paucity of specific and sensitive in vivo astrocyte biomarkers that can accurately measure preclinical AD. Measuring monoamine oxidase-B with neuroimaging and glial fibrillary acidic protein from bodily fluids are biomarkers that are currently available. Developing novel, more specific, and sensitive astrocyte biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial astrocytic functions in response to AD pathology. This review discusses astrocyte biomarkers in the context of AD.
Collapse
Affiliation(s)
- Stephen F Carter
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, Canada; Douglas Hospital Research Centre, Montreal, Canada; Montreal Neurological Institute, Montreal, Canada
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, Lausanne, Switzerland; Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, Bordeaux Cedex 33760, France
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Eduardo R Zimmer
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute (BraIns) of Rio Grande do Sul, Porto Alegre, Brazil; Website: www.zimmer-lab.org.
| |
Collapse
|
26
|
Carlstrom M, Montenegro MF. Therapeutic value of stimulating the nitrate-nitrite-nitric oxide pathway to attenuate oxidative stress and restore nitric oxide bioavailability in cardiorenal disease. J Intern Med 2019; 285:2-18. [PMID: 30039620 DOI: 10.1111/joim.12818] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disorders including hypertension and associated renal disease are major health problems affecting more than 1.5 billion people worldwide. Apart from nonmodifiable factors such as ageing, family history and gender, both sedentary lifestyle and unhealthy dietary habits are considered as major risk factors. The disorders are interrelated suggesting common pathological pathways. Mechanistically, oxidative stress and compromised function of the nitric oxide synthase (NOS) system leading to endothelial dysfunction and reduction in nitric oxide (NO) bioavailability have been widely implicated and associated with development and progression of disease. New strategies that correct this redox imbalance and increase NO bioactivity may have major clinical implications. The inorganic anions, nitrate and nitrite, are endogenously formed by oxidization of NOS-derived NO, but there are also high amounts of nitrate in our daily diet. In this regard, accumulated evidence over the past two decades demonstrates that these anions can be recycled back to NO and other bioactive nitrogen oxides, thus offering an attractive alternative strategy for therapeutic exploitation. In this review, we describe how dietary stimulation of the nitrate-nitrite-NO pathway affects cardiovascular and renal functions in health and disease via modulation of oxidative stress and NO bioavailability. Clinical studies addressing potential effects on the renal system are still limited, but blood pressure-lowering effects of nitrate supplementation have been demonstrated in healthy and hypertensive subjects as well as in patients with chronic kidney disease. However, larger clinical studies are warranted to reveal whether chronic nitrate treatment can slow-down the progression of cardiorenal disease and associated complications.
Collapse
Affiliation(s)
- M Carlstrom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - M F Montenegro
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Dehhaghi M, Tan V, Heng B, Mohammadipanah F, Guillemin GJ. Protective Effects of Myxobacterial Extracts on Hydrogen Peroxide-induced Toxicity on Human Primary Astrocytes. Neuroscience 2018; 399:1-11. [PMID: 30496822 DOI: 10.1016/j.neuroscience.2018.11.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/18/2018] [Accepted: 11/21/2018] [Indexed: 12/19/2022]
Abstract
Astrocytes, the main non-neuronal cells in the brain, have significant roles in the maintenance and survival of neurons. Oxidative stress has been implicated in various neurodegenerative disorders such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD). Myxobacteria produce a wide range of bioactive metabolites with notable structures and modes of action, which introduce them as potent natural product producers. In the present study, we evaluated the effects of myxobacterial extracts on hydrogen peroxide (H2O2)-mediated toxicity on primary human astrocytes. We showed that myxobacterial extracts could decrease the formation of reactive oxygen species (ROS), nitric oxide (NO) production, and cell death assessed by the release of lactate dehydrogenase (LDH). Myxobacterial extracts were also able to reduce the nitric oxide synthase (NOS) activity. The extracts reduced the oxidative effect of H2O2 on over-activation of poly (ADP-ribose) polymerase (PARP1), therefore preventing the cell death by restoring the NAD+ levels. In addition, myxobacterial extracts ameliorated the oxidative stress by increasing the glutathione level in cells. The overall results showed myxobacterial extracts, especially from the strains Archangium sp. UTMC 4070 and Cystobacter sp. UTMC 4073, were able to protect human primary astrocytes from oxidative stress.
Collapse
Affiliation(s)
- Mona Dehhaghi
- Departmentof Microbial Biotechnology, School of Biology and Centre of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran; NeuroinflammationGroup, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Vanessa Tan
- NeuroinflammationGroup, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Benjamin Heng
- NeuroinflammationGroup, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Fatemeh Mohammadipanah
- Departmentof Microbial Biotechnology, School of Biology and Centre of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.
| | - Gilles J Guillemin
- NeuroinflammationGroup, Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia.
| |
Collapse
|
28
|
Edwards TM, Hamlin HJ. Reproductive endocrinology of environmental nitrate. Gen Comp Endocrinol 2018; 265:31-40. [PMID: 29577898 DOI: 10.1016/j.ygcen.2018.03.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022]
Abstract
Nitrate is a widespread contaminant of aquatic ecosystems and drinking water. It is also broadly active in organismal physiology, and as such, has the potential to both enhance and disrupt normal physiological function. In animals, nitrate is a proposed endocrine disrupter that is converted in vivo to nitrite and nitric oxide. Nitric oxide, in particular, is a potent cell signaling molecule that participates in diverse biological pathways and events. Here, we review in vivo nitrate cycling and downstream mechanistic physiology, with an emphasis on reproductive outcomes. However, in many cases, the research produces contradictory results, in part because there is good evidence that nitrate follows a non-monotonic dose-response curve. This conundrum highlights an array of opportunities for scientists from different fields to collaborate for a full understanding of nitrate physiology. Opposing conclusions are especially likely when in vivo/in vitro, long term/short term, high dose/low dose, or hypoxia/normoxia studies are compared. We conclude that in vivo studies are most appropriate for testing an organism's integrated endocrine response to nitrate. Based on the limited available studies, there is a generalized trend that shorter term studies (less than 1 month) or studies involving low doses (≤5 mg/L NO3-N) cause steroid hormone levels to decline. Studies that last more than a month and/or involve higher, but still environmentally relevant, exposures (>50-100 mg/L NO3-N) cause steroid hormone levels to increase. Very high nitrate doses (>500 mg/L NO3-N) are cytotoxic in many species. Hypoxia and acidity are likely to intensify the effects of nitrate. For study design, degree of study animal reproductive maturity or activity is important, with immature/reproductively quiescent animals responding to nitrate differently, compared with reproductively active animals. A detailed table of studies is presented.
Collapse
Affiliation(s)
- Thea M Edwards
- Department of Biology, University of the South, Sewanee, TN, USA.
| | | |
Collapse
|
29
|
Carlström M, Lundberg JO, Weitzberg E. Mechanisms underlying blood pressure reduction by dietary inorganic nitrate. Acta Physiol (Oxf) 2018; 224:e13080. [PMID: 29694703 DOI: 10.1111/apha.13080] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/28/2018] [Accepted: 04/18/2018] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) importantly contributes to cardiovascular homeostasis by regulating blood flow and maintaining endothelial integrity. Conversely, reduced NO bioavailability is a central feature during natural ageing and in many cardiovascular disorders, including hypertension. The inorganic anions nitrate and nitrite are endogenously formed after oxidation of NO synthase (NOS)-derived NO and are also present in our daily diet. Knowledge accumulated over the past two decades has demonstrated that these anions can be recycled back to NO and other bioactive nitrogen oxides via serial reductions that involve oral commensal bacteria and various enzymatic systems. Intake of inorganic nitrate, which is predominantly found in green leafy vegetables and beets, has a variety of favourable cardiovascular effects. As hypertension is a major risk factor of morbidity and mortality worldwide, much attention has been paid to the blood pressure reducing effect of inorganic nitrate. Here, we describe how dietary nitrate, via stimulation of the nitrate-nitrite-NO pathway, affects various organ systems and discuss underlying mechanisms that may contribute to the observed blood pressure-lowering effect.
Collapse
Affiliation(s)
- M. Carlström
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - J. O. Lundberg
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - E. Weitzberg
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
30
|
Abstract
Nitric oxide (NO), generated from L-arginine and oxygen by NO synthases, is a pleiotropic signaling molecule involved in cardiovascular and metabolic regulation. More recently, an alternative pathway for the formation of this free radical has been explored. The inorganic anions nitrate (NO3-) and nitrite (NO2-), originating from dietary and endogenous sources, generate NO bioactivity in a process involving seemingly symbiotic oral bacteria and host enzymes in blood and tissues. The described cardio-metabolic effects of dietary nitrate from experimental and clinical studies include lowering of blood pressure, improved endothelial function, increased exercise performance, and reversal of metabolic syndrome, as well as antidiabetic effects. The mechanisms underlying the salutary metabolic effects of nitrate are being revealed and include interaction with mitochondrial respiration, activation of key metabolic regulatory pathways, and reduction of oxidative stress. Here we review the recent advances in the nitrate-nitrite-NO pathway, focusing on metabolic effects in health and disease.
Collapse
|
31
|
Hill RL, Kulbe JR, Singh IN, Wang JA, Hall ED. Synaptic Mitochondria are More Susceptible to Traumatic Brain Injury-induced Oxidative Damage and Respiratory Dysfunction than Non-synaptic Mitochondria. Neuroscience 2018; 386:265-283. [PMID: 29960045 DOI: 10.1016/j.neuroscience.2018.06.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/12/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) results in mitochondrial dysfunction and induction of lipid peroxidation (LP). Lipid peroxidation-derived neurotoxic aldehydes such as 4-HNE and acrolein bind to mitochondrial proteins, inducing additional oxidative damage and further exacerbating mitochondrial dysfunction and LP. Mitochondria are heterogeneous, consisting of both synaptic and non-synaptic populations. Synaptic mitochondria are reported to be more vulnerable to injury; however, this is the first study to characterize the temporal profile of synaptic and non-synaptic mitochondria following TBI, including investigation of respiratory dysfunction and oxidative damage to mitochondrial proteins between 3 and 120 h following injury. These results indicate that synaptic mitochondria are indeed the more vulnerable population, showing both more rapid and severe impairments than non-synaptic mitochondria. By 24 h, synaptic respiration is significantly impaired compared to synaptic sham, whereas non-synaptic respiration does not decline significantly until 48 h. Decreases in respiration are associated with increases in oxidative damage to synaptic and non-synaptic mitochondrial proteins at 48 h and 72 h, respectively. These results indicate that the therapeutic window for mitochondria-targeted pharmacological neuroprotectants to prevent respiratory dysfunction is shorter for the more vulnerable synaptic mitochondria than for the non-synaptic population.
Collapse
Affiliation(s)
- Rachel L Hill
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Jacqueline R Kulbe
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Indrapal N Singh
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Juan A Wang
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Edward D Hall
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States.
| |
Collapse
|
32
|
Modulation of Cellular Respiration by Endogenously Produced Nitric Oxide in Rat Hippocampal Slices. Methods Mol Biol 2018. [PMID: 29850995 DOI: 10.1007/978-1-4939-7831-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Nitric oxide (•NO) is an ubiquitous signaling molecule that participates in molecular processes associated with several neural phenomena ranging from memory formation to excitotoxicity. In the hippocampus, neuronal •NO production is coupled to the activation of NMDA type glutamate receptors. Cytochrome c oxidase has emerged as a novel target for •NO, which competes with O2 for binding to this mitochondrial complex. This reaction establishes •NO as a regulator of cellular metabolism and, possibly, mitochondrial production of reactive oxygen species which participate in cellular signaling. A major gap in the understanding of •NO bioactivity, namely, in the hippocampus, has been the lack of knowledge of its concentration dynamics. Here, we present a detailed description of the simultaneous recording of •NO and O2 concentration dynamics in rat hippocampal slices. Carbon fiber microelectrodes are fabricated and applied for real-time measurements of both gases in a system close to in vivo models. This approach allows for a better understanding of the current paradigm by which an intricate interplay between •NO and O2 regulates cellular respiration.
Collapse
|
33
|
Barros LF, Bolaños JP, Bonvento G, Bouzier-Sore AK, Brown A, Hirrlinger J, Kasparov S, Kirchhoff F, Murphy AN, Pellerin L, Robinson MB, Weber B. Current technical approaches to brain energy metabolism. Glia 2018; 66:1138-1159. [PMID: 29110344 PMCID: PMC5903992 DOI: 10.1002/glia.23248] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/14/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022]
Abstract
Neuroscience is a technology-driven discipline and brain energy metabolism is no exception. Once satisfied with mapping metabolic pathways at organ level, we are now looking to learn what it is exactly that metabolic enzymes and transporters do and when, where do they reside, how are they regulated, and how do they relate to the specific functions of neurons, glial cells, and their subcellular domains and organelles, in different areas of the brain. Moreover, we aim to quantify the fluxes of metabolites within and between cells. Energy metabolism is not just a necessity for proper cell function and viability but plays specific roles in higher brain functions such as memory processing and behavior, whose mechanisms need to be understood at all hierarchical levels, from isolated proteins to whole subjects, in both health and disease. To this aim, the field takes advantage of diverse disciplines including anatomy, histology, physiology, biochemistry, bioenergetics, cellular biology, molecular biology, developmental biology, neurology, and mathematical modeling. This article presents a well-referenced synopsis of the technical side of brain energy metabolism research. Detail and jargon are avoided whenever possible and emphasis is given to comparative strengths, limitations, and weaknesses, information that is often not available in regular articles.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, 5110466, Chile
| | - Juan P Bolaños
- Instituto de Biologia Funcional y Genomica-CSIC, Universidad de Salamanca, CIBERFES, Salamanca, 37007, Spain
| | - Gilles Bonvento
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Research Center (MIRCen), CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Anne-Karine Bouzier-Sore
- Centre de Résonance Magnétique des Systèmes Biologiques UMR 5536, CNRS-Université Bordeaux 146 rue Léo-Saignat, Bordeaux, France
| | - Angus Brown
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Johannes Hirrlinger
- Carl Ludwig Institute of Physiology, University of Leipzig, Liebigstr. 27, D-04103, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, D-37075, Germany
| | - Sergey Kasparov
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, BS8 1TD, United Kingdom
- Baltic Federal University, Kalinigrad, Russian Federation
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Building 48, Homburg, 66421, Germany
| | - Anne N Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093
| | - Luc Pellerin
- Département de Physiologie, 7 rue du Bugnon, Lausanne, CH1005, Switzerland
| | - Michael B Robinson
- Department of Pediatrics, and Department of Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc Natl Acad Sci U S A 2018; 115:5839-5848. [PMID: 29802228 DOI: 10.1073/pnas.1804932115] [Citation(s) in RCA: 697] [Impact Index Per Article: 99.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oxygen-derived free radicals and related oxidants are ubiquitous and short-lived intermediates formed in aerobic organisms throughout life. These reactive species participate in redox reactions leading to oxidative modifications in biomolecules, among which proteins and lipids are preferential targets. Despite a broad array of enzymatic and nonenzymatic antioxidant systems in mammalian cells and microbes, excess oxidant formation causes accumulation of new products that may compromise cell function and structure leading to cell degeneration and death. Oxidative events are associated with pathological conditions and the process of normal aging. Notably, physiological levels of oxidants also modulate cellular functions via homeostatic redox-sensitive cell signaling cascades. On the other hand, nitric oxide (•NO), a free radical and weak oxidant, represents a master physiological regulator via reversible interactions with heme proteins. The bioavailability and actions of •NO are modulated by its fast reaction with superoxide radical ([Formula: see text]), which yields an unusual and reactive peroxide, peroxynitrite, representing the merging of the oxygen radicals and •NO pathways. In this Inaugural Article, I summarize early and remarkable developments in free radical biochemistry and the later evolution of the field toward molecular medicine; this transition includes our contributions disclosing the relationship of •NO with redox intermediates and metabolism. The biochemical characterization, identification, and quantitation of peroxynitrite and its role in disease processes have concentrated much of our attention. Being a mediator of protein oxidation and nitration, lipid peroxidation, mitochondrial dysfunction, and cell death, peroxynitrite represents both a pathophysiologically relevant endogenous cytotoxin and a cytotoxic effector against invading pathogens.
Collapse
|
35
|
López-Gambero AJ, Martínez F, Salazar K, Cifuentes M, Nualart F. Brain Glucose-Sensing Mechanism and Energy Homeostasis. Mol Neurobiol 2018; 56:769-796. [PMID: 29796992 DOI: 10.1007/s12035-018-1099-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/25/2018] [Indexed: 01/02/2023]
Abstract
The metabolic and energy state of the organism depends largely on the availability of substrates, such as glucose for ATP production, necessary for maintaining physiological functions. Deregulation in glucose levels leads to the appearance of pathological signs that result in failures in the cardiovascular system and various diseases, such as diabetes, obesity, nephropathy, and neuropathy. Particularly, the brain relies on glucose as fuel for the normal development of neuronal activity. Regions adjacent to the cerebral ventricles, such as the hypothalamus and brainstem, exercise central control in energy homeostasis. These centers house nuclei of neurons whose excitatory activity is sensitive to changes in glucose levels. Determining the different detection mechanisms, the phenotype of neurosecretion, and neural connections involving glucose-sensitive neurons is essential to understanding the response to hypoglycemia through modulation of food intake, thermogenesis, and activation of sympathetic and parasympathetic branches, inducing glucagon and epinephrine secretion and other hypothalamic-pituitary axis-dependent counterregulatory hormones, such as glucocorticoids and growth hormone. The aim of this review focuses on integrating the current understanding of various glucose-sensing mechanisms described in the brain, thereby establishing a relationship between neuroanatomy and control of physiological processes involved in both metabolic and energy balance. This will advance the understanding of increasingly prevalent diseases in the modern world, especially diabetes, and emphasize patterns that regulate and stimulate intake, thermogenesis, and the overall synergistic effect of the neuroendocrine system.
Collapse
Affiliation(s)
- A J López-Gambero
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Málaga, Spain
| | - F Martínez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - K Salazar
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - M Cifuentes
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Málaga, Spain.
| | - F Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile. .,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile.
| |
Collapse
|
36
|
Gamal M, Moawad J, Rashed L, Morcos MA, Sharawy N. Possible involvement of tetrahydrobiopterin in the disturbance of redox homeostasis in sepsis - Induced brain dysfunction. Brain Res 2018; 1685:19-28. [PMID: 29428597 DOI: 10.1016/j.brainres.2018.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/04/2018] [Accepted: 02/05/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM Tetrahydrobiopterin (BH4) is an essential co-factor that regulates nitric oxide (NO) and reactive oxygen species (ROS) production by nitric oxide synthases (NOS). In this study, we evaluated the effects of sepsis on BH4 level and redox status in the brain by using the rat model of sepsis-induced by cecal ligation and puncture (CLP) and examined whether BH4 and/or acetyl-L-carnitine (ALC) could prevent the neuronal apoptosis and neurological changes induced by sepsis. MATERIAL AND METHOD Male albino rats were randomly and blindly divided into 8 groups: sham, sham + BH4, sham + ALC, sham +BH4+ ALC, CLP, CLP + BH4, CLP + ALC, and CLP+BH4+ ALC. We measured neurological indicators, brain levels of BH4, guanosine triphosphate cyclohydrolase (GTPCH), sepiapterin reductase (SR) and dihydropteridine reductase (DHPR) genes expression (Essential enzymes in BH4 biosynthesis and recycling pathways). We investigated also brain redox status and both endothelial and inducible NOS expressions. RESULTS Brain of septic rats demonstrated a reduced BH4 bioavailability, downregulation of BH4 synthetic enzymes, increased production of hydrogen peroxide and impaired antioxidant enzymes activities. Treatments with BH4 and/or ALC increased BH4 level, upregulated BH4 synthetic enzymes expressions, and attenuated oxidative-induced neuronal apoptosis. CONCLUSION Our results suggest that BH4 and/or ALC might protect the brain against oxidative stress induced neuronal apoptosis by restoring bioavailability of BH4 and upregulating of BH4 synthetic enzymes in the brain during sepsis.
Collapse
Affiliation(s)
- Maha Gamal
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Jackline Moawad
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Laila Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mary Attia Morcos
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nivin Sharawy
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt; Cairo University Hospitals, Cairo, Egypt.
| |
Collapse
|
37
|
Nitric Oxide and Mitochondrial Function in Neurological Diseases. Neuroscience 2018; 376:48-71. [DOI: 10.1016/j.neuroscience.2018.02.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/20/2018] [Accepted: 02/09/2018] [Indexed: 12/17/2022]
|
38
|
Blood Mononuclear Cell Mitochondrial Respiratory Chain Complex IV Activity Is Decreased in Multiple Sclerosis Patients: Effects of β-Interferon Treatment. J Clin Med 2018; 7:jcm7020036. [PMID: 29461488 PMCID: PMC5852452 DOI: 10.3390/jcm7020036] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/15/2018] [Accepted: 02/18/2018] [Indexed: 12/03/2022] Open
Abstract
Objectives: Evidence of mitochondrial respiratory chain (MRC) dysfunction and oxidative stress has been implicated in the pathophysiology of multiple sclerosis (MS). However, at present, there is no reliable low invasive surrogate available to evaluate mitochondrial function in these patients. In view of the particular sensitivity of MRC complex IV to oxidative stress, the aim of this study was to assess blood mononuclear cell (BMNC) MRC complex IV activity in MS patients and compare these results to age matched controls and MS patients on β-interferon treatment. Methods: Spectrophotometric enzyme assay was employed to measure MRC complex IV activity in blood mononuclear cell obtained multiple sclerosis patients and aged matched controls. Results: MRC Complex IV activity was found to be significantly decreased (p < 0.05) in MS patients (2.1 ± 0.8 k/nmol × 10−3; mean ± SD] when compared to the controls (7.2 ± 2.3 k/nmol × 10−3). Complex IV activity in MS patients on β-interferon (4.9 ± 1.5 k/nmol × 10−3) was not found to be significantly different from that of the controls. Conclusions: This study has indicated evidence of peripheral MRC complex IV deficiency in MS patients and has highlighted the potential utility of BMNCs as a potential means to evaluate mitochondrial function in this disorder. Furthermore, the reported improvement of complex IV activity may provide novel insights into the mode(s) of action of β-interferon.
Collapse
|
39
|
Inorganic nitrate alleviates the senescence-related decline in liver function. SCIENCE CHINA-LIFE SCIENCES 2018; 61:24-34. [DOI: 10.1007/s11427-017-9207-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/26/2017] [Indexed: 12/23/2022]
|
40
|
Parviainen L, Dihanich S, Anderson GW, Wong AM, Brooks HR, Abeti R, Rezaie P, Lalli G, Pope S, Heales SJ, Mitchison HM, Williams BP, Cooper JD. Glial cells are functionally impaired in juvenile neuronal ceroid lipofuscinosis and detrimental to neurons. Acta Neuropathol Commun 2017; 5:74. [PMID: 29041969 PMCID: PMC5645909 DOI: 10.1186/s40478-017-0476-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 09/23/2017] [Indexed: 11/18/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs or Batten disease) are a group of inherited, fatal neurodegenerative disorders of childhood. In these disorders, glial (microglial and astrocyte) activation typically occurs early in disease progression and predicts where neuron loss subsequently occurs. We have found that in the most common juvenile form of NCL (CLN3 disease or JNCL) this glial response is less pronounced in both mouse models and human autopsy material, with the morphological transformation of both astrocytes and microglia severely attenuated or delayed. To investigate their properties, we isolated glia and neurons from Cln3-deficient mice and studied their basic biology in culture. Upon stimulation, both Cln3-deficient astrocytes and microglia also showed an attenuated ability to transform morphologically, and an altered protein secretion profile. These defects were more pronounced in astrocytes, including the reduced secretion of a range of neuroprotective factors, mitogens, chemokines and cytokines, in addition to impaired calcium signalling and glutamate clearance. Cln3-deficient neurons also displayed an abnormal organization of their neurites. Most importantly, using a co-culture system, Cln3-deficient astrocytes and microglia had a negative impact on the survival and morphology of both Cln3-deficient and wildtype neurons, but these effects were largely reversed by growing mutant neurons with healthy glia. These data provide evidence that CLN3 disease astrocytes are functionally compromised. Together with microglia, they may play an active role in neuron loss in this disorder and can be considered as potential targets for therapeutic interventions.
Collapse
|
41
|
Understanding a role for hypoxia in lesion formation and location in the deep and periventricular white matter in small vessel disease and multiple sclerosis. Clin Sci (Lond) 2017; 131:2503-2524. [PMID: 29026001 DOI: 10.1042/cs20170981] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 08/01/2017] [Accepted: 08/15/2017] [Indexed: 12/28/2022]
Abstract
The deep and periventricular white matter is preferentially affected in several neurological disorders, including cerebral small vessel disease (SVD) and multiple sclerosis (MS), suggesting that common pathogenic mechanisms may be involved in this injury. Here we consider the potential pathogenic role of tissue hypoxia in lesion development, arising partly from the vascular anatomy of the affected white matter. Specifically, these regions are supplied by a sparse vasculature fed by long, narrow end arteries/arterioles that are vulnerable to oxygen desaturation if perfusion is reduced (as in SVD, MS and diabetes) or if the surrounding tissue is hypoxic (as in MS, at least). The oxygen crisis is exacerbated by a local preponderance of veins, as these can become highly desaturated 'sinks' for oxygen that deplete it from surrounding tissues. Additional haemodynamic deficiencies, including sluggish flow and impaired vasomotor reactivity and vessel compliance, further exacerbate oxygen insufficiency. The cells most vulnerable to hypoxic damage, including oligodendrocytes, die first, resulting in demyelination. Indeed, in preclinical models, demyelination is prevented if adequate oxygenation is maintained by raising inspired oxygen concentrations. In agreement with this interpretation, there is a predilection of lesions for the anterior and occipital horns of the lateral ventricles, namely regions located at arterial watersheds, or border zones, known to be especially susceptible to hypoperfusion and hypoxia. Finally, mitochondrial dysfunction due to genetic causes, as occurs in leucodystrophies or due to free radical damage, as occurs in MS, will compound any energy insufficiency resulting from hypoxia. Viewing lesion formation from the standpoint of tissue oxygenation not only reveals that lesion distribution is partly predictable, but may also inform new therapeutic strategies.
Collapse
|
42
|
de la Fuente C, Burke DG, Eaton S, Heales SJ. Inhibition of neuronal mitochondrial complex I or lysosomal glucocerebrosidase is associated with increased dopamine and serotonin turnover. Neurochem Int 2017; 109:94-100. [DOI: 10.1016/j.neuint.2017.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 01/31/2023]
|
43
|
Mitochondrial form, function and signalling in aging. Biochem J 2017; 473:3421-3449. [PMID: 27729586 DOI: 10.1042/bcj20160451] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 06/17/2016] [Indexed: 02/07/2023]
Abstract
Aging is often accompanied by a decline in mitochondrial mass and function in different tissues. Additionally, cell resistance to stress is frequently found to be prevented by higher mitochondrial respiratory capacity. These correlations strongly suggest mitochondria are key players in aging and senescence, acting by regulating energy homeostasis, redox balance and signalling pathways central in these processes. However, mitochondria display a wide array of functions and signalling properties, and the roles of these different characteristics are still widely unexplored. Furthermore, differences in mitochondrial properties and responses between tissues and cell types, and how these affect whole body metabolism are also still poorly understood. This review uncovers aspects of mitochondrial biology that have an impact upon aging in model organisms and selected mammalian cells and tissues.
Collapse
|
44
|
Effect of nitric oxide to axonal degeneration in multiple sclerosis via downregulating monocarboxylate transporter 1 in oligodendrocytes. Nitric Oxide 2017; 67:75-80. [PMID: 28392448 DOI: 10.1016/j.niox.2017.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS). Axonal degeneration, one of the main pathological characteristics of MS, is affected by nitric oxide (NO). In turn, NO induces mitochondrial dysfunction of neurons and glial cells. Inadequate glucose causes monocarboxylate transporter 1 (MCT1) to transfer lactate from oligodendrocytes (OLs) to neurons, which decreases MCT1 and results in energy substrate deficit (mainly lactate) in axons. The condition gradually leads to axonal degeneration. This study proposes that NO-induced MCT1 down-regulation in OLs may be involved in the pathological process of axonal degeneration, which eventually leads to MS.
Collapse
|
45
|
Morán J, Perez-Basterrechea M, Garrido P, Díaz E, Alonso A, Otero J, Colado E, González C. Effects of Estrogen and Phytoestrogen Treatment on an In Vitro Model of Recurrent Stroke on HT22 Neuronal Cell Line. Cell Mol Neurobiol 2017; 37:405-416. [PMID: 27059741 PMCID: PMC11482143 DOI: 10.1007/s10571-016-0372-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/01/2016] [Indexed: 01/01/2023]
Abstract
An increase of stroke incidence occurs in women with the decline of estrogen levels following menopause. This ischemic damage may recur, especially soon after the first insult has occurred. We evaluated the effects of estrogen and phytoestrogen treatment on an in vitro recurrent stroke model using the HT22 neuronal cell line. HT22 cells were treated with 17β-estradiol or genistein 1 h after the beginning of the first of two oxygen and glucose deprivation/reoxygenation (OGD/R) cycles. During the second OGD, there was a deterioration of some components of the electron transport chain, such as cytochrome c oxidase subunit 1 with a subsequent increase of reactive oxygen species (ROS) production. Accordingly, there was also an increase of apoptotic phenomena demonstrated by poly(ADP-ribose) polymerase 1 cleavage, Caspase-3 activity, and Annexin V levels. The recurrent ischemic injury also raised the hypoxia-inducible factor 1α and glucose transporter 1 levels, as well as the ratio between the lipidated and cytosolic forms of microtubule-associated protein 1A/1B-light chain 3 (LC3-II/LC3-I). We found a positive effect of estradiol and genistein treatment by partially preserving the impaired cell viability after the recurrent ischemic injury; however, this positive effect does not seem to be mediated neither by blocking apoptosis processes nor by decreasing ROS production. This work contribute to the better understanding of the molecular mechanisms triggered by recurrent ischemic damage in neuronal cells and, therefore, could help with the development of an effective treatment to minimize the consequences of this pathology.
Collapse
Affiliation(s)
- Javier Morán
- Department of Functional Biology. Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006, Oviedo, Spain
- Department of Clinical Neuroscience and Rehabilitation, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 9A, Box 440, 413 90, Gothenburg, Sweden
| | - Marcos Perez-Basterrechea
- Unit of Transplants, Cell Therapy and Regenerative Medicine, Central University Hospital of Asturias, Av. Roma s/n, 33011, Oviedo, Spain
| | - Pablo Garrido
- Department of Functional Biology. Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006, Oviedo, Spain
- Prostate Cancer Institute, National University of Ireland, Galway. Co, Galway, Ireland
| | - Elena Díaz
- Department of Functional Biology. Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006, Oviedo, Spain
| | - Ana Alonso
- Department of Functional Biology. Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006, Oviedo, Spain
| | - Jesús Otero
- Unit of Transplants, Cell Therapy and Regenerative Medicine, Central University Hospital of Asturias, Av. Roma s/n, 33011, Oviedo, Spain
| | - Enrique Colado
- Service of Hematology, Central University Hospital of Asturias, Av/Roma s/n, 33011, Oviedo, Spain
- Department of Laboratory Medicine, Central University Hospital of Asturias, Av/Roma s/n, 33011, Oviedo, Spain
| | - Celestino González
- Department of Functional Biology. Physiology Area, University of Oviedo, Av. Julián Clavería, No. 6, 33006, Oviedo, Spain.
| |
Collapse
|
46
|
Hohnholt MC, Blumrich EM, Waagepetersen HS, Dringen R. The tricarboxylic acid cycle activity in cultured primary astrocytes is strongly accelerated by the protein tyrosine kinase inhibitor tyrphostin 23. Neurochem Int 2016; 102:13-21. [PMID: 27894844 DOI: 10.1016/j.neuint.2016.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 11/28/2022]
Abstract
Tyrphostin 23 (T23) is a well-known inhibitor of protein tyrosine kinases and has been considered as potential anti-cancer drug. T23 was recently reported to acutely stimulate the glycolytic flux in primary cultured astrocytes. To investigate whether T23 also affects the tricarboxylic acid (TCA) cycle, we incubated primary rat astrocyte cultures with [U-13C]glucose in the absence or the presence of 100 μM T23 for 2 h and analyzed the 13C metabolite pattern. These incubation conditions did not compromise cell viability and confirmed that the presence of T23 doubled glycolytic lactate production. In addition, T23-treatment strongly increased the molecular carbon labeling of the TCA cycle intermediates citrate, succinate, fumarate and malate, and significantly increased the incorporation of 13C-labelling into the amino acids glutamate, glutamine and aspartate. These results clearly demonstrate that, in addition to glycolysis, also the mitochondrial TCA cycle is strongly accelerated after exposure of astrocytes to T23, suggesting that a protein tyrosine kinase may be involved in the regulation of the TCA cycle in astrocytes.
Collapse
Affiliation(s)
- Michaela C Hohnholt
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Eva-Maria Blumrich
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, PO. Box 330440, D-28334 Bremen, Germany; Centre for Environmental Research and Sustainable Technology, Leobener Strasse, D-28359 Bremen, Germany
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, PO. Box 330440, D-28334 Bremen, Germany; Centre for Environmental Research and Sustainable Technology, Leobener Strasse, D-28359 Bremen, Germany
| |
Collapse
|
47
|
Porta F, Takala J, Weikert C, Kaufmann P, Krahenbuhl S, Jakob SM. Effect of endotoxin, dobutamine and dopamine on muscle mitochondrial respiration in vitro. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519060120060601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Introduction: Mitochondrial respiration is impaired during endotoxemia. While catecholamines are frequently used in sepsis, their effects on mitochondrial function are controversial. We assessed effects of dobutamine and dopamine endotoxin on isolated muscle mitochondria. Materials and Methods : Sternocleidomastoid muscle mitochondria were isolated from six anesthetized pigs. Each sample was divided into six different groups. Three groups were incubated with endotoxin, three with vehicle. After 1 h, dopamine and dobutamine at final concentrations of 100 µM were added to the vehicle and endotoxin groups. After 2 h, state 3 and 4 respiration rates were determined for all mitochondrial complexes. Oxygen consumption was determined with a Clark-type electrode. Results: Endotoxin increased glutamate-dependent state 4 respiration from 9.3 ± 3.6 to 31.9 ± 9.1 ( P = 0.001) without affecting state 3 respiration. This reduced the efficiency of mitochondrial respiration (RCR; state 3/state 4, 9.9 ± 1.9 versus 3.6 ± 0.6; P < 0.001). The other complexes were unaffected. Catecholamine partially restored the endotoxin-induced increase in complex I state 4 respiration rate (31.9 ± 9.1 versus 17.1 ± 6.4 and 20.1 ± 12.2) after dopamine and dobutamine, respectively ( P = 0.007), and enhanced the ADP:O ratio ( P = 0.033). Conclusions: Dopamine and dobutamine enhanced the efficiency of mitochondrial respiration after short-term endotoxin exposure.
Collapse
Affiliation(s)
- Francesca Porta
- Department of Intensive Care Medicine, University Hospital Bern, Bern, Switzerland
| | - Jukka Takala
- Department of Intensive Care Medicine, University Hospital Bern, Bern, Switzerland
| | - Christian Weikert
- Department of Intensive Care Medicine, University Hospital Bern, Bern, Switzerland
| | - Priska Kaufmann
- Department of Clinical Pharmacology & Toxicology and Department of Research, University Hospital, Basel, Switzerland
| | - Stephan Krahenbuhl
- Department of Clinical Pharmacology & Toxicology and Department of Research, University Hospital, Basel, Switzerland,
| | - Stephan M. Jakob
- Department of Intensive Care Medicine, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
48
|
The neglected role of copper ions in wound healing. J Inorg Biochem 2016; 161:1-8. [DOI: 10.1016/j.jinorgbio.2016.02.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/19/2016] [Accepted: 02/10/2016] [Indexed: 12/30/2022]
|
49
|
Abstract
The molecular basis of migraine is still not completely understood. An impairment of mitochondrial oxidative metabolism might play a role in the pathophysiology of this disease, by influencing neuronal information processing. Biochemical assays of platelets and muscle biopsies performed in migraine sufferers have shown a decreased activity of the respiratory chain enzymes. Studies with phosphorus magnetic resonance spectroscopy (31P-MRS) have demonstrated an impairment of the brain oxidative energy metabolism both during and between migraine attacks. However, molecular genetic studies have not detected specific mitochondrial DNA (mtDNA) mutations in patients with migraine, although other studies suggest that particular genetic markers (i.e. neutral polymorphisms or secondary mtDNA mutations) might be present in some migraine sufferers. Further studies are still needed to clarify if migraine is associated with unidentified mutations on the mtDNA or on nuclear genes that code mitochondrial proteins. In this paper, we review morphological, biochemical, imaging and genetic studies which bear on the hypothesis that migraine may be related to mitochondrial dysfunction at least in some individuals.
Collapse
Affiliation(s)
- M Sparaco
- Department of Neurology and Headache Centre, Hospital G. Rummo Benevento, Benevento, Italy.
| | | | | | | | | |
Collapse
|
50
|
Quantification of Metabolic Rearrangements During Neural Stem Cells Differentiation into Astrocytes by Metabolic Flux Analysis. Neurochem Res 2016; 42:244-253. [DOI: 10.1007/s11064-016-1907-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 12/27/2022]
|