1
|
Lu W, Wen J. Metabolic reprogramming and astrocytes polarization following ischemic stroke. Free Radic Biol Med 2025; 228:197-206. [PMID: 39756488 DOI: 10.1016/j.freeradbiomed.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/28/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Astrocytes are critical for maintaining neuronal activity. Activation of astrocytes, occurs within minutes from ischemic stroke onset due to ischemic causes and subsequent inflammatory damage. Activated astrocytes, also known as reactive astrocytes, are divided into two different phenotypes: A1 (pro-inflammatory) and A2 (anti-inflammatory) astrocytes. A2 astrocytes support neuronal survival and promote tissue healing, while A1 astrocytes have neurotoxic effects. Thus, polarization of reactive astrocyte into A1 or A2 genotype is closely correlated with the development of cerebral ischemia/reperfusion (I/R) injury. Metabolic reprogramming is a process that various metabolic pathways upregulate in cells to balance energy, alter their phenotype, and produce building-block requirements. A1 and A2 astrocytes display different metabolic reprogramming, such as glycolysis, glutamate uptake, and glycogenolysis. Accumulating evidence suggested that manipulation of energy metabolism homeostasis can induce astrocytes to switch from A1 to A2 phenotype. This review disucss the potential factors in affecting astrocytic polarization, emphasizes metabolic reprogramming in reactive astrocytes within the pathophysiological context of cerebral I/R, and explores the relationship between metabolic reprogramming and astrocytic polarization. Importantly, we reveal that regulating metabolic reprogramming in reactive astrocytes may be a potential therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Yang HH, Chien WC, Liaw JJ, Yang CC, Chung CH, Huang SH, Huang YC, Wang BL, Chung RJ, Chen PC, Lin TT, Yu PC, Chen YJ. Impact of glycemic treatment and blood glucose monitoring on outcomes in patients with acute ischemic stroke without prior diabetes: a longitudinal cohort study. Diabetol Metab Syndr 2024; 16:302. [PMID: 39696458 DOI: 10.1186/s13098-024-01542-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVES To explore the short- and long-term effects of glycemic management-through glycemic treatment and blood glucose monitoring (BGM)-on stroke recurrence and mortality specifically in patients experiencing a first-ever ischemic stroke (FIS) with hyperglycemia (FISHG) who have not previously been diagnosed with diabetes mellitus (DM). METHODS We gathered data on patients who were registered on Taiwan's National Health Insurance Research Database from 2000 to 2015. We one-fold propensity-score-matched (by sex, age, and index date) 207,054 patients into 3 cohorts: those with FIS (1) without hyperglycemia, (2) hyperglycemia without glycemic treatment, and (3) hyperglycemia with glycemic treatment. We used Cox proportional hazard regression to evaluate the short- (within 1 year after FIS) and long-term (9.3 ± 8.6 years after FIS) prognostic effects of glycemic management on stroke recurrence and mortality of FISHG. RESULTS Stroke recurrence and mortality were significantly more likely in the patients with FISHG than their counterparts without hyperglycemia (p < 0.05). Under glycemic treatment, patients with FISHG demonstrated lower risk of mortality at every follow-up than those without (p < 0.001) but were not less likely to have stroke recurrence (p > 0.05). Integrating BGM with glycemic treatment in the FISHG cohort significantly reduced the risk of stroke recurrence compared to patients receiving only glycemic treatment at 1-month, 3-month, 6-month, and 1-year post-stroke follow-ups (adjusted hazard ratios = 0.84, 0.90, 0.88, and 0.92, respectively); additionally, this approach significantly decreased mortality risk at each post-stroke follow-up period (p < 0.05). CONCLUSIONS BGM combined with glycemic treatment significantly improves prognosis in patients with FISHG who have not been previously diagnosed with DM, reducing the risks of stroke recurrence and mortality.
Collapse
Affiliation(s)
- Hsi-Hsing Yang
- Department of Intensive Care Medicine, Chi-Mei Medical Center, Tainan, 71004, Taiwan
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan, 71005, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, Taipei, 11490, Taiwan.
- School of Public Health, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Jen-Jiuan Liaw
- School of Nursing, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chia-Chen Yang
- School of Nursing, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, Taipei, 11490, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Shi-Hao Huang
- Department of Medical Research, Tri-Service General Hospital, Taipei, 11490, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, 11490, Taiwan
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei, 10608, Taiwan
| | - Yao-Ching Huang
- Department of Medical Research, Tri-Service General Hospital, Taipei, 11490, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, 11490, Taiwan
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei, 10608, Taiwan
| | - Bing-Long Wang
- School of Public Health, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology (Taipei Tech), Taipei, 10608, Taiwan
| | - Peng-Ciao Chen
- School of Nursing, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Ting-Ti Lin
- School of Nursing, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Pi-Ching Yu
- Graduate Institute of Medicine, National Defense Medical Center, Taipei, 11490, Taiwan
- Cardiovascular Intensive Care Unit, Department of Critical Care Medicine, Far-Eastern Memorial Hospital, New Taipei City, 10602, Taiwan
| | - Yu-Ju Chen
- School of Nursing, National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
3
|
Kobzeva KA, Gurtovoy DE, Polonikov AV, Pokrovsky VM, Patrakhanov EA, Bushueva OY. Polymorphism in Genes Encoding HSP40 Family Proteins is Associated with Ischemic Stroke Risk and Brain Infarct Size: A Pilot Study. J Integr Neurosci 2024; 23:211. [PMID: 39735968 DOI: 10.31083/j.jin2312211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Heat shock proteins (HSPs) play a critical role in the molecular mechanisms of ischemic stroke (IS). A possible role for HSP40 family proteins in atherosclerosis progression has already been revealed; however, to date, molecular genetic studies on the involvement of genes encoding proteins of the HSP40 family in IS have not yet been carried out. AIM We sought to determine whether nine single nucleotide polymorphisms (SNPs) in genes encoding HSP40 family proteins (DNAJB1, DNAJB2, DNAJA1, DNAJA2, DNAJA3 and DNAJC7) are associated with the risk and clinical features of IS. METHODS Using TaqMan-based polymerase chain reaction (PCR) and the MassArray-4 system, DNA samples of 2551 Russians - 1306 IS patients and 1245 healthy individuals - were genotyped. RESULTS SNP rs2034598 DNAJA2 decreased the risk of IS exclusively in male patients (odds ratio = 0.81, 95% confidence interval 0.78-0.98, p = 0.028); rs7189628 DNAJA2 increased the brain infarct size (p = 0.04); and rs6500605 DNAJA3 lowered the age of onset of IS (p = 0.03). SNPs rs10448231 DNAJA1, rs7189628 DNAJA2, rs4926222 DNAJB1 and rs2034598 DNAJA2 were involved in the strongest epistatic interactions linked to IS; SNP rs10448231 DNAJA1 is characterised by the most essential mono-effect (2.96% of IS entropy); all of the top SNP-SNP interaction models included the pairwise combination rs7189628 DNAJA2×rs4926222 DNAJB1, which was found to be a key factor determining susceptibility to IS. In interactions with the studied SNPs, smoking was found to have multidirectional effects (synergism, antagonism or additive effect) and the strongest mono-effect (3.47% of IS entropy), exceeding the mono-effects of rs6500605 DNAJA3, rs10448231 DNAJA1, rs2034598 DNAJA2, rs7189628 DNAJA2 and rs4926222 DNAJB1, involved in the best G×E models and determining 0.03%-0.73% of IS entropy. CONCLUSIONS We are the first to discover polymorphisms in genes encoding HSP40 family proteins as a major risk factor for IS and its clinical manifestations. The comprehensive bioinformatics analysis revealed molecular mechanisms, underscoring their significance in the pathogenesis of IS, primarily reflecting the regulation of heat stress, proteostasis and cellular signalling.
Collapse
Affiliation(s)
- Ksenia A Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Denis E Gurtovoy
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Alexey V Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| | - Vladimir M Pokrovsky
- Laboratory of Genetic Technologies and Gene Editing for Biomedicine and Veterinary Medicine, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny A Patrakhanov
- Laboratory of Genetic Technologies and Gene Editing for Biomedicine and Veterinary Medicine, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olga Y Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| |
Collapse
|
4
|
Brookshier A, Lyden P. Differential vulnerability among cell types in the neurovascular unit: Description and mechanisms. J Cereb Blood Flow Metab 2024:271678X241299960. [PMID: 39520113 PMCID: PMC11563522 DOI: 10.1177/0271678x241299960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Currently, successful preclinical cerebroprotective agents fail to translate effectively into clinical practice suggesting the need for a comprehensive evaluation of all aspects of brain function. Selective vulnerability refers to the specific regional response of the brain following global ischemia, with observed patterns of vulnerability attributed to the distribution of neuronal subtypes and the functions of respective brain regions. Conversely, the concept of differential vulnerability pertains to the cell-type-specific reactions to cerebral ischemia, dictated by the biological characteristics of individual cells. This review aims to explore these vulnerability hypotheses and elucidate potential underlying cellular mechanisms.
Collapse
Affiliation(s)
- Allison Brookshier
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute of the Keck School of Medicine of USC, Los Angeles, USA
| | - Patrick Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute of the Keck School of Medicine of USC, Los Angeles, USA
- Department of Neurology, Keck School of Medicine of USC, Los Angeles, USA
| |
Collapse
|
5
|
Sun M, Ruan X, Zhou Z, Huo Y, Liu M, Liu S, Cao J, Liu YH, Zhang X, Ma YL, Mi W. Effect of intranasal insulin on perioperative cognitive function in older adults: a randomized, placebo-controlled, double-blind clinical trial. Age Ageing 2024; 53:afae188. [PMID: 39216470 DOI: 10.1093/ageing/afae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 06/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Postoperative cognitive impairment are common neural complications in older surgical patients and exacerbate the burden of medical care on families and society. METHODS A total of 140 older patients who were scheduled for elective orthopaedic surgery or pancreatic surgery with general anaesthesia were randomly assigned to Group S or Group I with a 1:1 allocation. Patients in Group S and Group I received intranasal administration of 400 μL of normal saline or 40 IU/400 μL of insulin, respectively, once daily from 5 minutes before anaesthesia induction until 3 days postoperatively. Perioperative cognitive function was assessed using the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment-Basic (MoCA-B) at 1 day before and 3 days after surgery and postoperative delirium (POD) incidence was assessed using the 3-minute Diagnostic Interview for CAM (3D-CAM) on postoperative days 1-3. Serum levels of interleukin-6 (IL-6), tumour necrosis factor α (TNF-α), S100-β and C-reactive protein (CRP) were measured on the first day after surgery. RESULTS Insulin treatment significantly increased postoperative MMSE and MoCA-B scores in group I than in group S (P < 0.001, P = 0.001, respectively), decreased the incidence of POD within the 3-day postoperative period in Group I than in Group S (10.9% vs 26.6%, P = 0.024), and inhibited postoperative IL-6 and S100-β levels in Group I compared to Group S (P = 0.034, P = 0.044, respectively). CONCLUSIONS Intranasal insulin administration is thus suggested as a potential therapy to improve postoperative cognition in older patients undergoing surgery. However, a more standardized multi-centre, large-sample study is needed to further validate these results.
Collapse
Affiliation(s)
- Miao Sun
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
- Department of Anesthesiology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xianghan Ruan
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Zhikang Zhou
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Yuting Huo
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Min Liu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Siyuan Liu
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Jiangbei Cao
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Yan-Hong Liu
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Xiaoying Zhang
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Yu-Long Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| | - Weidong Mi
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100730, China
| |
Collapse
|
6
|
Guo H, Li Y, Wang S, Yang Y, Xu T, Zhao J, Wang J, Zuo W, Wang P, Zhao G, Wang H, Hou W, Dong H, Cai Y. Dysfunction of astrocytic glycophagy exacerbates reperfusion injury in ischemic stroke. Redox Biol 2024; 74:103234. [PMID: 38861834 PMCID: PMC11215420 DOI: 10.1016/j.redox.2024.103234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
Glycophagy has evolved from an alternative glycogen degradation pathway into a multifaceted pivot to regulate cellular metabolic hemostasis in peripheral tissues. However, the pattern of glycophagy in the brain and its potential therapeutic impact on ischemic stroke remain unknown. Here, we observed that the dysfunction of astrocytic glycophagy was caused by the downregulation of the GABA type A receptor-associated protein like 1 (GABARAPL1) during reperfusion in ischemic stroke patients and mice. PI3K-Akt pathway activation is involved in driving GABARAPL1 downregulation during cerebral reperfusion. Moreover, glycophagy dysfunction-induced glucosamine deficiency suppresses the nuclear translocation of specificity protein 1 and TATA binding protein, the transcription factors for GABARAPL1, by decreasing their O-GlcNAcylation levels, and accordingly feedback inhibits GABARAPL1 in astrocytes during reperfusion. Restoring astrocytic glycophagy by overexpressing GABARAPL1 decreases DNA damage and oxidative injury in astrocytes and improves the survival of surrounding neurons during reperfusion. In addition, a hypocaloric diet in the acute phase after cerebral reperfusion can enhance astrocytic glycophagic flux and accelerate neurological recovery. In summary, glycophagy in the brain links autophagy, metabolism, and epigenetics together, and glycophagy dysfunction exacerbates reperfusion injury after ischemic stroke.
Collapse
Affiliation(s)
- Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yumeng Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongheng Yang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tiantian Xu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianshuai Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jin Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pengju Wang
- The State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guangchao Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huaning Wang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yanhui Cai
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Foley TD, Huang WC, Petsche EA, Fleming ER, Hornickle JC. Protein vicinal thiols as intrinsic probes of brain redox states in health, aging, and ischemia. Metab Brain Dis 2024; 39:929-940. [PMID: 38848024 PMCID: PMC11233328 DOI: 10.1007/s11011-024-01370-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/03/2024] [Indexed: 07/10/2024]
Abstract
The nature of brain redox metabolism in health, aging, and disease remains to be fully established. Reversible oxidations, to disulfide bonds, of closely spaced (vicinal) protein thiols underlie the catalytic maintenance of redox homeostasis by redoxin enzymes, including thioredoxin peroxidases (peroxiredoxins), and have been implicated in redox buffering and regulation. We propose that non-peroxidase proteins containing vicinal thiols that are responsive to physiological redox perturbations may serve as intrinsic probes of brain redox metabolism. Using redox phenylarsine oxide (PAO)-affinity chromatography, we report that PAO-binding vicinal thiols on creatine kinase B and alpha-enolase from healthy rat brains were preferentially oxidized compared to other selected proteins, including neuron-specific (gamma) enolase, under conditions designed to trap in vivo protein thiol redox states. Moreover, measures of the extents of oxidations of vicinal thiols on total protein, and on creatine kinase B and alpha-enolase, showed that vicinal thiol-linked redox states were stable over the lifespan of rats and revealed a transient reductive shift in these redox couples following decapitation-induced global ischemia. Finally, formation of disulfide-linked complexes between peroxiredoxin-2 and brain proteins was demonstrated on redox blots, supporting a link between protein vicinal thiol redox states and the peroxidase activities of peroxiredoxins. The implications of these findings with respect to underappreciated aspects of brain redox metabolism in health, aging, and ischemia are discussed.
Collapse
Affiliation(s)
- Timothy D Foley
- Biochemistry Program, Department of Chemistry, University of Scranton, Scranton, PA, 18510, USA.
| | - Wen C Huang
- Biochemistry Program, Department of Chemistry, University of Scranton, Scranton, PA, 18510, USA
| | - Emily A Petsche
- Biochemistry Program, Department of Chemistry, University of Scranton, Scranton, PA, 18510, USA
| | - Emily R Fleming
- Biochemistry Program, Department of Chemistry, University of Scranton, Scranton, PA, 18510, USA
| | - James C Hornickle
- Biochemistry Program, Department of Chemistry, University of Scranton, Scranton, PA, 18510, USA
| |
Collapse
|
8
|
Rae CD, Baur JA, Borges K, Dienel G, Díaz-García CM, Douglass SR, Drew K, Duarte JMN, Duran J, Kann O, Kristian T, Lee-Liu D, Lindquist BE, McNay EC, Robinson MB, Rothman DL, Rowlands BD, Ryan TA, Scafidi J, Scafidi S, Shuttleworth CW, Swanson RA, Uruk G, Vardjan N, Zorec R, McKenna MC. Brain energy metabolism: A roadmap for future research. J Neurochem 2024; 168:910-954. [PMID: 38183680 PMCID: PMC11102343 DOI: 10.1111/jnc.16032] [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: 05/27/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
Although we have learned much about how the brain fuels its functions over the last decades, there remains much still to discover in an organ that is so complex. This article lays out major gaps in our knowledge of interrelationships between brain metabolism and brain function, including biochemical, cellular, and subcellular aspects of functional metabolism and its imaging in adult brain, as well as during development, aging, and disease. The focus is on unknowns in metabolism of major brain substrates and associated transporters, the roles of insulin and of lipid droplets, the emerging role of metabolism in microglia, mysteries about the major brain cofactor and signaling molecule NAD+, as well as unsolved problems underlying brain metabolism in pathologies such as traumatic brain injury, epilepsy, and metabolic downregulation during hibernation. It describes our current level of understanding of these facets of brain energy metabolism as well as a roadmap for future research.
Collapse
Affiliation(s)
- Caroline D. Rae
- School of Psychology, The University of New South Wales, NSW 2052 & Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karin Borges
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Gerald Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Carlos Manlio Díaz-García
- Department of Biochemistry and Molecular Biology, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska, USA
| | - João M. N. Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, & Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, Baltimore, Maryland, USA
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dasfne Lee-Liu
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Región Metropolitana, Chile
| | - Britta E. Lindquist
- Department of Neurology, Division of Neurocritical Care, Gladstone Institute of Neurological Disease, University of California at San Francisco, San Francisco, California, USA
| | - Ewan C. McNay
- Behavioral Neuroscience, University at Albany, Albany, New York, USA
| | - Michael B. Robinson
- Departments of Pediatrics and System Pharmacology & Translational Therapeutics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas L. Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Benjamin D. Rowlands
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | - Joseph Scafidi
- Department of Neurology, Kennedy Krieger Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susanna Scafidi
- Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine Albuquerque, Albuquerque, New Mexico, USA
| | - Raymond A. Swanson
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Gökhan Uruk
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology—Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mary C. McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Markussen KH, Corti M, Byrne BJ, Kooi CWV, Sun RC, Gentry MS. The multifaceted roles of the brain glycogen. J Neurochem 2024; 168:728-743. [PMID: 37554056 PMCID: PMC10901277 DOI: 10.1111/jnc.15926] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023]
Abstract
Glycogen is a biologically essential macromolecule that is directly involved in multiple human diseases. While its primary role in carbohydrate storage and energy metabolism in the liver and muscle is well characterized, recent research has highlighted critical metabolic and non-metabolic roles for glycogen in the brain. In this review, the emerging roles of glycogen homeostasis in the healthy and diseased brain are discussed with a focus on advancing our understanding of the role of glycogen in the brain. Innovative technologies that have led to novel insights into glycogen functions are detailed. Key insights into how cellular localization impacts neuronal and glial function are discussed. Perturbed glycogen functions are observed in multiple disorders of the brain, including where it serves as a disease driver in the emerging category of neurological glycogen storage diseases (n-GSDs). n-GSDs include Lafora disease (LD), adult polyglucosan body disease (APBD), Cori disease, Glucose transporter type 1 deficiency syndrome (G1D), GSD0b, and late-onset Pompe disease (PD). They are neurogenetic disorders characterized by aberrant glycogen which results in devastating neurological and systemic symptoms. In the most severe cases, rapid neurodegeneration coupled with dementia results in death soon after diagnosis. Finally, we discuss current treatment strategies that are currently being developed and have the potential to be of great benefit to patients with n-GSD. Taken together, novel technologies and biological insights have resulted in a renaissance in brain glycogen that dramatically advanced our understanding of both biology and disease. Future studies are needed to expand our understanding and the multifaceted roles of glycogen and effectively apply these insights to human disease.
Collapse
Affiliation(s)
- Kia H. Markussen
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, USA
| | - Manuela Corti
- Department of Pediatrics, Powell Gene Therapy Center, College of Medicine, University of Florida, USA
| | - Barry J. Byrne
- Department of Pediatrics, Powell Gene Therapy Center, College of Medicine, University of Florida, USA
| | - Craig W. Vander Kooi
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida
- Lafora Epilepsy Cure Initiative
| | - Ramon C. Sun
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida
- Lafora Epilepsy Cure Initiative
| | - Matthew S. Gentry
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida
- Lafora Epilepsy Cure Initiative
| |
Collapse
|
10
|
Brewer MK, Torres P, Ayala V, Portero-Otin M, Pamplona R, Andrés-Benito P, Ferrer I, Guinovart JJ, Duran J. Glycogen accumulation modulates life span in a mouse model of amyotrophic lateral sclerosis. J Neurochem 2024; 168:744-759. [PMID: 37401737 PMCID: PMC10764643 DOI: 10.1111/jnc.15906] [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: 02/28/2023] [Revised: 05/30/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of motor neurons in the spinal cord. Glial cells, including astrocytes and microglia, have been shown to contribute to neurodegeneration in ALS, and metabolic dysfunction plays an important role in the progression of the disease. Glycogen is a soluble polymer of glucose found at low levels in the central nervous system that plays an important role in memory formation, synaptic plasticity, and the prevention of seizures. However, its accumulation in astrocytes and/or neurons is associated with pathological conditions and aging. Importantly, glycogen accumulation has been reported in the spinal cord of human ALS patients and mouse models. In the present work, using the SOD1G93A mouse model of ALS, we show that glycogen accumulates in the spinal cord and brainstem during symptomatic and end stages of the disease and that the accumulated glycogen is associated with reactive astrocytes. To study the contribution of glycogen to ALS progression, we generated SOD1G93A mice with reduced glycogen synthesis (SOD1G93A GShet mice). SOD1G93A GShet mice had a significantly longer life span than SOD1G93A mice and showed lower levels of the astrocytic pro-inflammatory cytokine Cxcl10, suggesting that the accumulation of glycogen is associated with an inflammatory response. Supporting this, inducing an increase in glycogen synthesis reduced life span in SOD1G93A mice. Altogether, these results suggest that glycogen in reactive astrocytes contributes to neurotoxicity and disease progression in ALS.
Collapse
Affiliation(s)
- M. Kathryn Brewer
- Institute for Research in Biomedicine of Barcelona (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Pascual Torres
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida-IRB Lleida, Lleida, Spain
| | - Victòria Ayala
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida-IRB Lleida, Lleida, Spain
| | - Manuel Portero-Otin
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida-IRB Lleida, Lleida, Spain
| | - Reinald Pamplona
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida-IRB Lleida, Lleida, Spain
| | - Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Hospitalet de Llobregat, Spain
| | - Joan J. Guinovart
- Institute for Research in Biomedicine of Barcelona (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine of Barcelona (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
11
|
Madai S, Kilic P, Schmidt RM, Bas-Orth C, Korff T, Büttner M, Klinke G, Poschet G, Marti HH, Kunze R. Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics 2024; 14:2856-2880. [PMID: 38773968 PMCID: PMC11103502 DOI: 10.7150/thno.88223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 04/08/2024] [Indexed: 05/24/2024] Open
Abstract
Cell metabolism reprogramming to sustain energy production, while reducing oxygen and energy consuming processes is crucially important for the adaptation to hypoxia/ischemia. Adaptive metabolic rewiring is controlled by hypoxia-inducible factors (HIFs). Accumulating experimental evidence indicates that timely activation of HIF in brain-resident cells improves the outcome from acute ischemic stroke. However, the underlying molecular mechanisms are still incompletely understood. Thus, we investigated whether HIF-dependent metabolic reprogramming affects the vulnerability of brain-resident cells towards ischemic stress. Methods: We used genetic and pharmacological approaches to activate HIF in the murine brain in vivo and in primary neurons and astrocytes in vitro. Numerous metabolomic approaches and molecular biological techniques were applied to elucidate potential HIF-dependent effects on the central carbon metabolism of brain cells. In animal and cell models of ischemic stroke, we analysed whether HIF-dependent metabolic reprogramming influences the susceptibility to ischemic injury. Results: Neuron-specific gene ablation of prolyl-4-hydroxylase domain 2 (PHD2) protein, negatively regulating the protein stability of HIF-α in an oxygen dependent manner, reduced brain injury and functional impairment of mice after acute stroke in a HIF-dependent manner. Accordingly, PHD2 deficient neurons showed an improved tolerance towards ischemic stress in vitro, which was accompanied by enhanced HIF-1-mediated glycolytic lactate production through pyruvate dehydrogenase kinase-mediated inhibition of the pyruvate dehydrogenase. Systemic treatment of mice with roxadustat, a low-molecular weight pan-PHD inhibitor, not only increased the abundance of numerous metabolites of the central carbon and amino acid metabolism in murine brain, but also ameliorated cerebral tissue damage and sensorimotor dysfunction after acute ischemic stroke. In neurons and astrocytes roxadustat provoked a HIF-1-dependent glucose metabolism reprogramming including elevation of glucose uptake, glycogen synthesis, glycolytic capacity, lactate production and lactate release, which enhanced the ischemic tolerance of astrocytes, but not neurons. We found that strong activation of HIF-1 in neurons by non-selective inhibition of all PHD isoenzymes caused a HIF-1-dependent upregulation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 redirecting glucose-6-phosphate from pentose phosphate pathway (PPP) to the glycolysis pathway. This was accompanied by a reduction of NADPH production in the PPP, which further decreased the low intrinsic antioxidant reserve of neurons, making them more susceptible to ischemic stress. Nonetheless, in organotypic hippocampal cultures with preserved neuronal-glial interactions roxadustat decreased the neuronal susceptibility to ischemic stress, which was largely prevented by restricting glycolytic energy production through lactate transport blockade. Conclusion: Collectively, our results indicate that HIF-1-mediated metabolic reprogramming alleviates the intrinsic vulnerability of brain-resident cells to ischemic stress.
Collapse
Affiliation(s)
- Sarah Madai
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Pinar Kilic
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Rolf M. Schmidt
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Michael Büttner
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Glynis Klinke
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Hugo H. Marti
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Reiner Kunze
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
12
|
Hirayama Y, Le HPN, Hashimoto H, Ishii I, Koizumi S, Anzai N. Preconditioning-Induced Facilitation of Lactate Release from Astrocytes Is Essential for Brain Ischemic Tolerance. eNeuro 2024; 11:ENEURO.0494-23.2024. [PMID: 38604775 PMCID: PMC11064122 DOI: 10.1523/eneuro.0494-23.2024] [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: 11/26/2023] [Revised: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
A sublethal ischemic episode [termed preconditioning (PC)] protects neurons in the brain against a subsequent severe ischemic injury. This phenomenon is known as brain ischemic tolerance and has received much attention from researchers because of its robust neuroprotective effects. We have previously reported that PC activates astrocytes and subsequently upregulates P2X7 receptors, thereby leading to ischemic tolerance. However, the downstream signals of P2X7 receptors that are responsible for PC-induced ischemic tolerance remain unknown. Here, we show that PC-induced P2X7 receptor-mediated lactate release from astrocytes has an indispensable role in this event. Using a transient focal cerebral ischemia model caused by middle cerebral artery occlusion, extracellular lactate levels during severe ischemia were significantly increased in mice who experienced PC; this increase was dependent on P2X7 receptors. In addition, the intracerebroventricular injection of lactate protected against cerebral ischemic injury. In in vitro experiments, although stimulation of astrocytes with the P2X7 receptor agonist BzATP had no effect on the protein levels of monocarboxylate transporter (MCT) 1 and MCT4 (which are responsible for lactate release from astrocytes), BzATP induced the plasma membrane translocation of these MCTs via their chaperone CD147. Importantly, CD147 was increased in activated astrocytes after PC, and CD147-blocking antibody abolished the PC-induced facilitation of astrocytic lactate release and ischemic tolerance. Taken together, our findings suggest that astrocytes induce ischemic tolerance via P2X7 receptor-mediated lactate release.
Collapse
Affiliation(s)
- Yuri Hirayama
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Ha Pham Ngoc Le
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Itsuko Ishii
- Division of Pharmacy, Chiba University Hospital, Chiba 260-8677, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
13
|
Yang C, Wang H, Shao M, Chu F, He Y, Chen X, Fan J, Chen J, Cai Q, Wu C. Brain-Type Glycogen Phosphorylase (PYGB) in the Pathologies of Diseases: A Systematic Review. Cells 2024; 13:289. [PMID: 38334681 PMCID: PMC10854662 DOI: 10.3390/cells13030289] [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: 11/23/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 02/10/2024] Open
Abstract
Glycogen metabolism is a form of crucial metabolic reprogramming in cells. PYGB, the brain-type glycogen phosphorylase (GP), serves as the rate-limiting enzyme of glycogen catabolism. Evidence is mounting for the association of PYGB with diverse human diseases. This review covers the advancements in PYGB research across a range of diseases, including cancer, cardiovascular diseases, metabolic diseases, nervous system diseases, and other diseases, providing a succinct overview of how PYGB functions as a critical factor in both physiological and pathological processes. We present the latest progress in PYGB in the diagnosis and treatment of various diseases and discuss the current limitations and future prospects of this novel and promising target.
Collapse
Affiliation(s)
- Caiting Yang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Haojun Wang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Miaomiao Shao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Fengyu Chu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Yuyu He
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Xiaoli Chen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Jiahui Fan
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Jingwen Chen
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| | - Qianqian Cai
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (C.Y.); (H.W.); (F.C.); (Y.H.); (X.C.); (J.F.); (J.C.)
| |
Collapse
|
14
|
Wang Q, Qin Y, Ma J, Zhou K, Xia G, Li Y, Xie L, Afful RG, Lan Q, Huo X, Zou J, Yang H. An early warning indicator of mortality risk in patients with COVID-19: the neutrophil extracellular traps/neutrophilic segmented granulocyte ratio. Front Immunol 2024; 15:1287132. [PMID: 38348024 PMCID: PMC10859410 DOI: 10.3389/fimmu.2024.1287132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Background Neutrophil extracellular traps (NETs) play a key role in thrombus formation in patients with coronavirus disease 2019 (COVID-19). However, the existing detection and observation methods for NETs are limited in their ability to provide quantitative, convenient, and accurate descriptions of in situ NETs. Therefore, establishing a quantitative description of the relationship between NETs and thrombosis remains a challenge. Objective We employed morphological observations of blood cells and statistical analyses to investigate the correlation between the NETs/neutrophilic segmented granulocyte ratio and mortality risk in patients with COVID-19. Methods Peripheral blood samples were collected from 117 hospitalized patients with COVID-19 between November 2022 and February 2023, and various blood cell parameters were measured. Two types of smudge cells were observed in the blood and counted: lymphatic and neutral smudge cells. Statistical data analysis was used to establish COVID-19 mortality risk assessment indicators. Results Morphological observations of neutrophilic smudge cells revealed swelling, eruption, and NETs formation in the neutrophil nuclei. Subsequently, the NETs/neutrophilic segmented granulocyte ratio (NNSR) was calculated. A high concentration of NETs poses a fatal risk for thrombus formation in patients. Statistical analysis indicated that a high NNSR was more suitable for evaluating the risk of death in patients with COVID-19 compared to elevated fibrinogen (FIB) and D-dimer (DD) levels. Conclusion Observing blood cell morphology is an effective method for the detection of NETs, NNSR are important markers for revealing the mortality risk of patients with COVID-19.
Collapse
Affiliation(s)
- Qiong Wang
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Yu Qin
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jingyun Ma
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Kehao Zhou
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Guiping Xia
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ya Li
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Li Xie
- School of Internet of Things Engineering, Jiangnan University, Wuxi, China
| | - Richmond Godwin Afful
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Qian Lan
- School of Internet of Things Engineering, Jiangnan University, Wuxi, China
| | - Xingyu Huo
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jian Zou
- The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Hailin Yang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
15
|
Gong L, Liang J, Xie L, Zhang Z, Mei Z, Zhang W. Metabolic Reprogramming in Gliocyte Post-cerebral Ischemia/ Reperfusion: From Pathophysiology to Therapeutic Potential. Curr Neuropharmacol 2024; 22:1672-1696. [PMID: 38362904 PMCID: PMC11284719 DOI: 10.2174/1570159x22666240131121032] [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: 08/21/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/17/2024] Open
Abstract
Ischemic stroke is a leading cause of disability and death worldwide. However, the clinical efficacy of recanalization therapy as a preferred option is significantly hindered by reperfusion injury. The transformation between different phenotypes of gliocytes is closely associated with cerebral ischemia/ reperfusion injury (CI/RI). Moreover, gliocyte polarization induces metabolic reprogramming, which refers to the shift in gliocyte phenotype and the overall transformation of the metabolic network to compensate for energy demand and building block requirements during CI/RI caused by hypoxia, energy deficiency, and oxidative stress. Within microglia, the pro-inflammatory phenotype exhibits upregulated glycolysis, pentose phosphate pathway, fatty acid synthesis, and glutamine synthesis, whereas the anti-inflammatory phenotype demonstrates enhanced mitochondrial oxidative phosphorylation and fatty acid oxidation. Reactive astrocytes display increased glycolysis but impaired glycogenolysis and reduced glutamate uptake after CI/RI. There is mounting evidence suggesting that manipulation of energy metabolism homeostasis can induce microglial cells and astrocytes to switch from neurotoxic to neuroprotective phenotypes. A comprehensive understanding of underlying mechanisms and manipulation strategies targeting metabolic pathways could potentially enable gliocytes to be reprogrammed toward beneficial functions while opening new therapeutic avenues for CI/RI treatment. This review provides an overview of current insights into metabolic reprogramming mechanisms in microglia and astrocytes within the pathophysiological context of CI/RI, along with potential pharmacological targets. Herein, we emphasize the potential of metabolic reprogramming of gliocytes as a therapeutic target for CI/RI and aim to offer a novel perspective in the treatment of CI/RI.
Collapse
Affiliation(s)
- Lipeng Gong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Junjie Liang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Letian Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhanwei Zhang
- Department of Neurosurgery, First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410007, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei 443002, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
16
|
Song J, Zaidi SAA, He L, Zhang S, Zhou G. Integrative Analysis of Machine Learning and Molecule Docking Simulations for Ischemic Stroke Diagnosis and Therapy. Molecules 2023; 28:7704. [PMID: 38067435 PMCID: PMC10707570 DOI: 10.3390/molecules28237704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Due to the narrow therapeutic window and high mortality of ischemic stroke, it is of great significance to investigate its diagnosis and therapy. We employed weighted gene coexpression network analysis (WGCNA) to ascertain gene modules related to stroke and used the maSigPro R package to seek the time-dependent genes in the progression of stroke. Three machine learning algorithms were further employed to identify the feature genes of stroke. A nomogram model was built and applied to evaluate the stroke patients. We analyzed single-cell RNA sequencing (scRNA-seq) data to discern microglia subclusters in ischemic stroke. The RNA velocity, pseudo time, and gene set enrichment analysis (GSEA) were performed to investigate the relationship of microglia subclusters. Connectivity map (CMap) analysis and molecule docking were used to screen a therapeutic agent for stroke. A nomogram model based on the feature genes showed a clinical net benefit and enabled an accurate evaluation of stroke patients. The RNA velocity and pseudo time analysis showed that microglia subcluster 0 would develop toward subcluster 2 within 24 h from stroke onset. The GSEA showed that the function of microglia subcluster 0 was opposite to that of subcluster 2. AZ_628, which screened from CMap analysis, was found to have lower binding energy with Mmp12, Lgals3, Fam20c, Capg, Pkm2, Sdc4, and Itga5 in microglia subcluster 2 and maybe a therapeutic agent for the poor development of microglia subcluster 2 after stroke. Our study presents a nomogram model for stroke diagnosis and provides a potential molecule agent for stroke therapy.
Collapse
Affiliation(s)
- Jingwei Song
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
| | - Liangge He
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
| | - Shuai Zhang
- Brain Research Centre, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, China; (J.S.); (S.A.A.Z.); (L.H.)
- Lungene Biotech Ltd., Shenzhen 518060, China
- Senotherapeutics Ltd., Hangzhou 311100, China
| |
Collapse
|
17
|
Bar S, Wilson KA, Hilsabeck TA, Alderfer S, Dammer EB, Burton JB, Shah S, Holtz A, Carrera EM, Beck JN, Chen JH, Kauwe G, Tracy TE, Seyfried NT, Schilling B, Ellerby LM, Kapahi P. Neuronal Glycogen Breakdown Mitigates Tauopathy via Pentose Phosphate Pathway-Mediated Oxidative Stress Reduction. RESEARCH SQUARE 2023:rs.3.rs-3526342. [PMID: 37986935 PMCID: PMC10659530 DOI: 10.21203/rs.3.rs-3526342/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Tauopathies encompass a range of neurodegenerative disorders, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD). Unfortunately, current treatment approaches for tauopathies have yielded limited success, underscoring the pressing need for novel therapeutic strategies. We observed distinct signatures of impaired glycogen metabolism in the Drosophila brain of the tauopathy model and the brain of AD patients, indicating a link between tauopathies and glycogen metabolism. We demonstrate that the breakdown of neuronal glycogen by activating glycogen phosphorylase (GlyP) ameliorates the tauopathy phenotypes in flies and induced pluripotent stem cell (iPSC) derived neurons from FTD patients. We observed that glycogen breakdown redirects the glucose flux to the pentose phosphate pathway to alleviate oxidative stress. Our findings uncover a critical role for increased GlyP activity in mediating the neuroprotection benefit of dietary restriction (DR) through the cAMP-mediated protein kinase A (PKA) activation. Our studies identify impaired glycogen metabolism as a key hallmark for tauopathies and offer a promising therapeutic target in tauopathy treatment.
Collapse
Affiliation(s)
- Sudipta Bar
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| | | | | | | | - Eric B. Dammer
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory University, School of Medicine Core Labs, Atlanta, GA 30322, USA
| | | | - Samah Shah
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| | - Anja Holtz
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| | | | | | - Jackson H Chen
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| | - Tara E. Tracy
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA 94947, USA
| |
Collapse
|
18
|
Xu S, Li S, Yan Z, Wang Y, Zhang L. Development and Validation of a UHPLC-MS/MS Method for the Quantification of a Novel PYGB Inhibitor in Plasma: Application to Pharmacokinetic Studies. Molecules 2023; 28:6995. [PMID: 37836837 PMCID: PMC10574475 DOI: 10.3390/molecules28196995] [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: 08/11/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
In previous studies, we reported compound 1 (5-chloro-N-(4-oxo-2,2-dipropyl-3,4-dihydro-2H-benzo[e][1,3]oxazin-6-yl)-1H-indole-2-carboxamide) as a novel PYGB inhibitor, and found that it had better anti-ischemic brain injury activity. In this study, we established and validated a novel UHPLC-MS/MS method for the quantitative determination of compound 1 in plasma, then applied the method to study the pharmacokinetic parameters and brain tissue distribution of compound 1 in SD (Sprague-Dawley) rats after intravenous administration. The experimental results showed that the method met the validation requirements set by the US FDA in terms of linearity, accuracy, precision, and stability. The validated method was then used for pharmacokinetic studies in rat plasma, and it was found that compound 1 exhibited linear pharmacokinetic characteristics when administered in the dose range of 0.8-3.2 mg/kg. Finally, we also conducted a brief preliminary investigation of the brain tissue distribution of compound 1 in rats after injection and found that the brain tissue concentrations at 0.25 h and 2 h of administration were 440 ± 19.1 ng/kg and 111 ± 23.9 ng/kg, respectively. Additionally, the CBrain/CPlasma ratio was 0.112 ± 0.0185 and 0.112 ± 0.0292, respectively. These results indicated that compound 1 was able to cross the blood-brain barrier. This study provides important support for the application of compound 1 in ischemic brain injury diseases.
Collapse
Affiliation(s)
| | | | | | | | - Liying Zhang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China; (S.X.); (S.L.); (Z.Y.)
| |
Collapse
|
19
|
Clarkson-Paredes C, Karl MT, Popratiloff A, Miller RH. A unique cell population expressing the Epithelial-Mesenchymal Transition-transcription factor Snail moderates microglial and astrocyte injury responses. PNAS NEXUS 2023; 2:pgad334. [PMID: 37901440 PMCID: PMC10612478 DOI: 10.1093/pnasnexus/pgad334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023]
Abstract
Insults to the central nervous system (CNS) elicit common glial responses including microglial activation evidenced by functional, morphological, and phenotypic changes, as well as astrocyte reactions including hypertrophy, altered process orientation, and changes in gene expression and function. However, the cellular and molecular mechanisms that initiate and modulate such glial response are less well-defined. Here we show that an adult cortical lesion generates a population of ultrastructurally unique microglial-like cells that express Epithelial-Mesenchymal Transcription factors including Snail. Knockdown of Snail with antisense oligonucleotides results in a postinjury increase in activated microglial cells, elevation in astrocyte reactivity with increased expression of C3 and phagocytosis, disruption of astrocyte junctions and neurovascular structure, increases in neuronal cell death, and reduction in cortical synapses. These changes were associated with alterations in pro-inflammatory cytokine expression. By contrast, overexpression of Snail through microglia-targeted an adeno-associated virus (AAV) improved many of the injury characteristics. Together, our results suggest that the coordination of glial responses to CNS injury is partly mediated by epithelial-mesenchymal transition-factors (EMT-Fsl).
Collapse
Affiliation(s)
- Cheryl Clarkson-Paredes
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
- Nanofabrication and Imaging Center, The George Washington University, 800 22nd Street NW, Washington, DC 20052, USA
| | - Molly T Karl
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
| | - Anastas Popratiloff
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
- Nanofabrication and Imaging Center, The George Washington University, 800 22nd Street NW, Washington, DC 20052, USA
| | - Robert H Miller
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
| |
Collapse
|
20
|
Guo S, Qian C, Li W, Zeng Z, Cai J, Luo Y. Modulation of Neuroinflammation: Advances in Roles and Mechanisms of the IL-33/ST2 Axis Involved in Ischemic Stroke. Neuroimmunomodulation 2023; 30:226-236. [PMID: 37729881 PMCID: PMC10614518 DOI: 10.1159/000533984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/02/2023] [Indexed: 09/22/2023] Open
Abstract
Interleukin (IL)-33 was initially recognized as a constituent of the IL-1 cytokine family in 2005. It exerts pleiotropic effects by regulating immune responses via its binding to the receptor ST2 (IL-33R). The IL-33/ST2 pathway has been linked to several inflammatory disorders. In human and rodents, the broad expression of IL-33 in spinal cord tissues and brain indicates its central nervous system-specific functions. Growing evidence supports the protective effects of the IL-33/ST2 pathway in ischemic stroke, along with a better understanding of the underlying mechanisms. IL-33 plays a crucial role in the regulation of the release of inflammatory molecules from glial cells in response to neuropathological lesions. Moreover, IL-33/ST2-mediated neuroprotection following cerebral ischemia may be linked to T-cell function, specifically regulatory T cells. Soluble ST2 (sST2) acts as a decoy receptor in the IL-33/ST2 axis, blocking IL-33 signaling through the membrane ST2 receptor. sST2 has also been identified as a potential inflammatory biomarker of ischemic stroke. Targeting sST2 specifically to eliminate its inhibition of the protective IL-33/ST2 pathway in ischemic brain tissues is a promising approach for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Shuang Guo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chengli Qian
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenfeng Li
- Department of Clinical Medicine, The Second Clinical College, Wuhan University, Wuhan, China
| | - Zhikun Zeng
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junlong Cai
- Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Wang Y, Li S, Yan Z, Zhang L. Computational Insights into Novel Inhibitor N-(3-( tert-Butylcarbamoyl)-4-methoxyphenyl)-indole and Ingliforib Specific against GP Isoenzyme Dimers Interaction Mechanism. Molecules 2023; 28:4909. [PMID: 37446571 DOI: 10.3390/molecules28134909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
The high conservation of the three subtypes of glycogen phosphorylase (GP) presents significant challenges for specific inhibitor studies targeting GP. Our prior screening revealed that compound 1 exhibited unequal inhibitory activity against the three GP subtypes, with a noticeable effect against brain GP (PYGB). The commercially available ingliforib demonstrated potent inhibitory activity specifically against liver GP (PYGL). To guide the further design and screening of high-specificity inhibitors, the possible reasons for the differential inhibitory activity of two compounds against different GP subtypes were analyzed, with ingliforib as a reference, through molecular docking and molecular dynamics simulations. Initially, the study predicted the binding modes of ligands with the three GP receptor subtypes using molecular docking. Subsequently, this was validated by molecular dynamics experiments, and possible amino acid residues that had important interactions were explored. The strong correlation between the calculated interaction free energies and experimental inhibitory activity implied the reasonable binding conformations of the compounds. These findings offer insight into the different inhibitory activity of compound 1 and ingliforib against all three GP subtypes and provide guidance for the design of specific target molecules that regulate subtype selectivity.
Collapse
Affiliation(s)
- Youde Wang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Shuai Li
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Zhiwei Yan
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Liying Zhang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| |
Collapse
|
22
|
Yan Z, Li S, Wang Y, Guo Y, Zhang L. Protective effects of a novel glycogen phosphorylase inhibitor against cerebral ischemia-reperfusion injury in mice. Future Med Chem 2023; 15:587-597. [PMID: 37097106 DOI: 10.4155/fmc-2023-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Aim: To evaluate the effects of a novel glycogen phosphorylase inhibitor (NGPI) on cerebral ischemia-reperfusion injury (CIRI). Methods: Cerebral ischemia was induced in mice using a modified bilateral common carotid artery ligation model. To assess the effects of NGPI against CIRI, mice which had been administered with different doses of NGPI (1.25, 2.5, 5 mg/kg/day) for 7 days before the injury were evaluated for infarct volume, the apoptosis level of brain tissue, integrity of brain tissue and oxidative stress level. Results: NGPI effectively improved the infarct area, apoptosis of neurons, integrity of brain tissue and oxidative stress level of mice with CIRI. Conclusion: NGPI could effectively improve CIRI and deserves further study.
Collapse
Affiliation(s)
- Zhiwei Yan
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei, 067000, China
| | - Shuai Li
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei, 067000, China
| | - Youde Wang
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei, 067000, China
| | - Yachun Guo
- Department of Pathogen Biology, Chengde Medical University, Chengde, Hebei, 067000, China
| | - Liying Zhang
- Key Laboratory of Traditional Chinese Medicine Research & Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei, 067000, China
| |
Collapse
|
23
|
Guo H, Li Y, Hou W, Cai Y. Brain Glycogen: An Angel or a Devil for Ischemic Stroke? Neurosci Bull 2023; 39:690-694. [PMID: 36562984 PMCID: PMC10073389 DOI: 10.1007/s12264-022-01006-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/18/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yumeng Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yanhui Cai
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
24
|
Sex-Dimorphic Glucocorticoid Receptor Regulation of Hypothalamic Primary Astrocyte Glycogen Metabolism: Interaction with Norepinephrine. NEUROGLIA (BASEL, SWITZERLAND) 2022; 3:144-157. [PMID: 36685006 PMCID: PMC9850496 DOI: 10.3390/neuroglia3040010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Astrocyte glycogen is a critical metabolic variable that impacts hypothalamic control of glucostasis. Glucocorticoid hormones regulate peripheral glycogen, but their effects on hypothalamic glycogen are not known. A hypothalamic astrocyte primary culture model was used to investigate the premise that glucocorticoids impose sex-dimorphic independent and interactive control of glycogen metabolic enzyme protein expression and glycogen accumulation. The glucocorticoid receptor (GR) agonist dexamethasone (DEX) down-regulated glycogen synthase (GS), glycogen phosphorylase (GP)-brain type (GPbb), and GP-muscle type (GPmm) proteins in glucose-supplied male astrocytes, but enhanced these profiles in female. The catecholamine neurotransmitter norepinephrine (NE) did not alter these proteins, but amplified DEX inhibition of GS and GPbb in male or abolished GR stimulation of GPmm in female. In both sexes, DEX and NE individually increased glycogen content, but DEX attenuated the magnitude of noradrenergic stimulation. Glucoprivation suppressed GS, GPbb, and GPmm in male, but not female astrocytes, and elevated or diminished glycogen in these sexes, respectively. Glucose-deprived astrocytes exhibit GR-dependent induced glycogen accumulation in both sexes, and corresponding loss (male) or attenuation (female) of noradrenergic-dependent glycogen build-up. Current evidence for GR augmentation of hypothalamic astrocyte glycogen content in each sex, yet divergent effects on glycogen enzyme proteins infers that glucocorticoids may elicit opposite adjustments in glycogen turnover in each sex. Results document GR modulation of NE stimulation of glycogen accumulation in the presence (male and female) or absence (female) of glucose. Outcomes provide novel proof that astrocyte energy status influences the magnitude of GR and NE signal effects on glycogen mass.
Collapse
|
25
|
Newton J, Pushie M, Sylvain N, Hou H, Weese Maley S, Kelly M. Sex differences in the mouse photothrombotic stroke model investigated with X-ray fluorescence microscopy and Fourier transform infrared spectroscopic imaging. IBRO Neurosci Rep 2022; 13:127-135. [PMID: 35989697 PMCID: PMC9386104 DOI: 10.1016/j.ibneur.2022.07.006] [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: 04/22/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Stroke is a leading cause of death and disability around the world. To date, the majority of pre-clinical research has been performed using male lab animals and results are commonly generalized to both sexes. In clinical stoke cases females have a higher incidence of ischemic stroke and poorer outcomes, compared to males. Best practices for improving translatability of findings for stroke, encourage the use of both sexes in studies. Since estrogen and progesterone have recognized neuroprotective effects, it is important to compare the size, severity and biochemical composition of the brain tissue following stroke in female and male animal models. In this study a photothrombotic focal stroke was induced in male and female mice. Vaginal secretions were collected twice daily to track the stage of estrous. Mice were euthanized at 24 h post-stroke. Histological staining, Fourier transform infrared imaging and X-ray fluorescence imaging were performed to better define the size and metabolic markers in the infarct core and surrounding penumbra. Our results show while the female mice had a significantly lower body mass than males, the cross-sectional area of the brain and the size of infarct and penumbra were not significantly different between the groups. In addition to the general expected sex-linked differences of altered NADH levels between males and females, estrus females had significantly elevated glycogen in the penumbra compared with males and total phosphorus levels were noted to be higher in the penumbra of estrus females. Elevated glycogen reserves in the tissue bordering the infarct core in females may present alternatives for improved functional recovery in females in the early post-stroke phase.
Collapse
Affiliation(s)
- J.M. Newton
- Department of Surgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - M.J. Pushie
- Department of Surgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - N.J. Sylvain
- Department of Surgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
- Clinical Trial Support Unit, College of Medicine, University of Saskatchewan, SK S7N 0W8, Canada
| | - H. Hou
- Department of Surgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
| | - S. Weese Maley
- Clinical Trial Support Unit, College of Medicine, University of Saskatchewan, SK S7N 0W8, Canada
| | - M.E. Kelly
- Department of Surgery, College of Medicine, University of Saskatchewan, SK S7N 5E5, Canada
- Corresponding author.
| |
Collapse
|
26
|
The Mitochondrial Enzyme 17βHSD10 Modulates Ischemic and Amyloid-β-Induced Stress in Primary Mouse Astrocytes. eNeuro 2022; 9:ENEURO.0040-22.2022. [PMID: 36096650 PMCID: PMC9536859 DOI: 10.1523/eneuro.0040-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022] Open
Abstract
Severe brain metabolic dysfunction and amyloid-β accumulation are key hallmarks of Alzheimer's disease (AD). While astrocytes contribute to both pathologic mechanisms, the role of their mitochondria, which is essential for signaling and maintenance of these processes, has been largely understudied. The current work provides the first direct evidence that the mitochondrial metabolic switch 17β-hydroxysteroid dehydrogenase type 10 (17βHSD10) is expressed and active in murine astrocytes from different brain regions. While it is known that this protein is overexpressed in the brains of AD patients, we found that 17βHSD10 is also upregulated in astrocytes exposed to amyloidogenic and ischemic stress. Importantly, such catalytic overexpression of 17βHSD10 inhibits mitochondrial respiration during increased energy demand. This observation contrasts with what has been found in neuronal and cancer model systems, which suggests astrocyte-specific mechanisms mediated by the protein. Furthermore, the catalytic upregulation of the enzyme exacerbates astrocytic damage, reactive oxygen species (ROS) generation and mitochondrial network alterations during amyloidogenic stress. On the other hand, 17βHSD10 inhibition through AG18051 counters most of these effects. In conclusion, our data represents novel insights into the role of astrocytic mitochondria in metabolic and amyloidogenic stress with implications of 17βHSD10 in multiple neurodegenerative mechanisms.
Collapse
|
27
|
Huang Y, Li S, Wang Y, Yan Z, Guo Y, Zhang L. A Novel 5-Chloro-N-phenyl-1H-indole-2-carboxamide Derivative as Brain-Type Glycogen Phosphorylase Inhibitor: Potential Therapeutic Effect on Cerebral Ischemia. Molecules 2022; 27:molecules27196333. [PMID: 36234871 PMCID: PMC9572471 DOI: 10.3390/molecules27196333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Brain-type glycogen phosphorylase inhibitors are potential new drugs for treating ischemic brain injury. In our previous study, we reported compound 1 as a novel brain-type glycogen phosphorylase inhibitor with cardioprotective properties. We also found that compound 1 has high blood–brain barrier permeability through the ADMET prediction website. In this study, we deeply analyzed the protective effect of compound 1 on hypoxic-ischemic brain injury, finding that compound 1 could alleviate the hypoxia/reoxygenation (H/R) injury of astrocytes by improving cell viability and reducing LDH leakage rate, intracellular glucose content, and post-ischemic ROS level. At the same time, compound 1 could reduce the level of ATP in brain cells after ischemia, improve cellular energy metabolism, downregulate the degree of extracellular acidification, and improve metabolic acidosis. It could also increase the level of mitochondrial aerobic energy metabolism during brain cell reperfusion, reduce anaerobic glycolysis, and inhibit apoptosis and the expression of apoptosis-related proteins. The above results indicated that compound 1 is involved in the regulation of glucose metabolism, can control cell apoptosis, and has protective and potential therapeutic effects on cerebral ischemia-reperfusion injury, which provides a new reference and possibility for the development of novel drugs for the treatment of ischemic brain injury.
Collapse
Affiliation(s)
- Yatao Huang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Shuai Li
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Youde Wang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Zhiwei Yan
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Yachun Guo
- Department of Pathogen Biology, Chengde Medical University, Chengde 067000, China
- Correspondence: (Y.G.); (L.Z.); Tel.: +86-0314-229-1000 (L.Z.)
| | - Liying Zhang
- Laboratory of Traditional Chinese Medicine Research and Development of Hebei Province, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde 067000, China
- Correspondence: (Y.G.); (L.Z.); Tel.: +86-0314-229-1000 (L.Z.)
| |
Collapse
|
28
|
Wang S, Zhang Z, Wang J, Ma L, Zhao J, Wang J, Fang Z, Hou W, Guo H. Neuronal GPER Participates in Genistein-Mediated Neuroprotection in Ischemic Stroke by Inhibiting NLRP3 Inflammasome Activation in Ovariectomized Female Mice. Mol Neurobiol 2022; 59:5024-5040. [PMID: 35661323 DOI: 10.1007/s12035-022-02894-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/18/2022] [Indexed: 11/29/2022]
Abstract
Estrogen replacement therapy (ERT) is potentially beneficial for the prevention and treatment of postmenopausal cerebral ischemia but inevitably increases the risk of cerebral hemorrhage and breast cancer when used for a long period of time. Genistein, a natural phytoestrogen, has been reported to contribute to the recovery of postmenopausal ischemic stroke with reduced risks. However, the underlying mechanism of genistein-mediated neuroprotection remains unclear. We reported that genistein exerted significant neuroprotective effects by enhancing the expression of neuronal G protein-coupled estrogen receptor (GPER) in the ischemic penumbra after cerebral reperfusion in ovariectomized (OVX) mice, and this effect was achieved through GPER-mediated inhibition of nod-like receptor protein 3 (NLRP3) inflammasome activation. In addition, we found that peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α) was the pivotal molecule that participated in GPER-mediated inhibition of NLRP3 inflammasome activation in OVX mice after ischemia/reperfusion (I/R) injury. Our data suggest that the neuronal GPER/PGC-1α pathway plays an important role in genistein-mediated neuroprotection against I/R injury in OVX mice.
Collapse
Affiliation(s)
- Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhen Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jin Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lina Ma
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jianshuai Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jiajia Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zongping Fang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
29
|
Xie Y, Kuan AT, Wang W, Herbert ZT, Mosto O, Olukoya O, Adam M, Vu S, Kim M, Tran D, Gómez N, Charpentier C, Sorour I, Lacey TE, Tolstorukov MY, Sabatini BL, Lee WCA, Harwell CC. Astrocyte-neuron crosstalk through Hedgehog signaling mediates cortical synapse development. Cell Rep 2022; 38:110416. [PMID: 35196485 PMCID: PMC8962654 DOI: 10.1016/j.celrep.2022.110416] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/04/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
Neuron-glia interactions play a critical role in the regulation of synapse formation and circuit assembly. Here we demonstrate that canonical Sonic hedgehog (Shh) pathway signaling in cortical astrocytes acts to coordinate layer-specific synaptic connectivity. We show that the Shh receptor Ptch1 is expressed by cortical astrocytes during development and that Shh signaling is necessary and sufficient to promote the expression of genes involved in regulating synaptic development and layer-enriched astrocyte molecular identity. Loss of Shh in layer V neurons reduces astrocyte complexity and coverage by astrocytic processes in tripartite synapses; conversely, cell-autonomous activation of Shh signaling in astrocytes promotes cortical excitatory synapse formation. Furthermore, Shh-dependent genes Lrig1 and Sparc distinctively contribute to astrocyte morphology and synapse formation. Together, these results suggest that Shh secreted from deep-layer cortical neurons acts to specialize the molecular and functional features of astrocytes during development to shape circuit assembly and function.
Collapse
Affiliation(s)
- Yajun Xie
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron T Kuan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wengang Wang
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Olivia Mosto
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Olubusola Olukoya
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Manal Adam
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Steve Vu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Minsu Kim
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Diana Tran
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Nicolás Gómez
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Claire Charpentier
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ingie Sorour
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tiara E Lacey
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Y Tolstorukov
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Bernardo L Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wei-Chung Allen Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Zhu Y, Fan Z, Zhao Q, Li J, Cai G, Wang R, Liang Y, Lu N, Kang J, Luo D, Tao H, Li Y, Huang J, Wu S. Brain-Type Glycogen Phosphorylase Is Crucial for Astrocytic Glycogen Accumulation in Chronic Social Defeat Stress-Induced Depression in Mice. Front Mol Neurosci 2022; 14:819440. [PMID: 35140588 PMCID: PMC8820374 DOI: 10.3389/fnmol.2021.819440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/30/2021] [Indexed: 01/12/2023] Open
Abstract
Astrocytic glycogen plays an important role in brain energy metabolism. However, the contribution of glycogen metabolism to stress-induced depression remains unclear. Chronic social defeat stress was used to induce depression-like behaviors in mice, assessed with behavioral tests. Glycogen concentration in the medial prefrontal cortex (mPFC) and the expression of key enzymes of the glycogen metabolism were investigated using Western blots, immunofluorescent staining, electron microscopy, and biochemical assays. Stereotaxic surgery and viral-mediated gene transfer were applied to knockdown or overexpress brain-type glycogen phosphorylase (PYGB) in the mPFC. The glycogen content increased in the mPFC after stress. Glycogenolytic dysfunction due to inactivation of PYGB was responsible for glycogen accumulation. Behavioral tests on astrocyte-specific PYGB overexpression mice showed that augmenting astrocytic PYGB reduces susceptibility to depression when compared with stress-susceptible mice. Conversely, PYGB genetic down-regulation in the mPFC was sufficient to induce glycogen accumulation and depression-like behaviors. Furthermore, PYGB overexpression in the mPFC decreases susceptibility to depression, at least partially by rescuing glycogen phosphorylase activity to maintain glycogen metabolism homeostasis during stress. These findings indicate that (1) glycogen accumulation occurs in mice following stress and (2) glycogenolysis reprogramming leads to glycogen accumulation in astrocytes and PYGB contributes to stress-induced depression-like behaviors. Pharmacological tools acting on glycogenolysis might constitute a promising therapy for depression.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Ze Fan
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Military Stomatology, Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Qiuying Zhao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Jiaqi Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Guohong Cai
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Rui Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Yi Liang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Naining Lu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Junjun Kang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Danlei Luo
- Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huiren Tao
- Department of Spine Surgery, Shenzhen University General Hospital, Shenzhen, China
| | - Yan Li
- Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yan Li,
| | - Jing Huang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
- Jing Huang,
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
- Shengxi Wu,
| |
Collapse
|
31
|
Wei H, Zhen L, Wang S, Zhang Y, Wang K, Jia P, Zhang Y, Wu Z, Yang Q, Hou W, Lv J, Zhang P. De novo Lipogenesis in Astrocytes Promotes the Repair of Blood-Brain Barrier after Transient Cerebral Ischemia Through Interleukin-33. Neuroscience 2022; 481:85-98. [PMID: 34822949 DOI: 10.1016/j.neuroscience.2021.11.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/07/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022]
Abstract
Astrocytes experience significant metabolic shifts in the "sensitive period" of neurological function recovery following cerebral ischemia. However, the changes in astrocyte lipid metabolism and their implications for neurological recovery remain unknown. In the present study, we employed a mouse middle cerebral artery occlusion model to investigate the changes in de novo lipogenesis and interleukin-33 (IL-33) production in astrocytes and elucidate their role in blood-brain barrier (BBB) repair in the subacute phase of cerebral ischemia. Neurological behavior evaluation was used to assess functional changes in mice. Pharmacological inhibition and astrocyte-specific downregulation of fatty acid synthase (FASN) were used to evaluate the role of de novo lipogenesis in brain injury. Intracerebroventricular administration of recombinant IL-33 was performed to study the contribution of IL-33 to BBB disruption. Extravasation of Evans blue dye, dextran and IgG were used to assess BBB integrity. Western blotting of tight junction proteins ZO-1, Occludin, and Claudin-5 were performed at defined time points to evaluate changes in BBB. It was found that de novo lipogenesis was activated, and IL-33 production increased in astrocytes at the subacute stage of cerebral ischemia injury. Inhibition of lipogenesis in astrocytes decreased IL-33 production in the peri-infarct area, deteriorated BBB damage and interfered with neurological recovery. In addition, supplementation of IL-33 alleviated BBB destruction and improved neurological recovery worsened by lipogenesis inhibition. These findings indicate that astrocyte lipogenesis increases the production of IL-33 in the peri-infarct area, which promotes BBB repair in the subacute phase of cerebral ischemia injury and improves long-term functional recovery.
Collapse
Affiliation(s)
- Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Luming Zhen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yuanyuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Zhixin Wu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qianzi Yang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jianrui Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
32
|
Astrocytic glycogen mobilization participates in salvianolic acid B-mediated neuroprotection against reperfusion injury after ischemic stroke. Exp Neurol 2021; 349:113966. [PMID: 34973964 DOI: 10.1016/j.expneurol.2021.113966] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/22/2021] [Accepted: 12/23/2021] [Indexed: 01/01/2023]
Abstract
Astrocytic glycogen serves as an important glucose reserve, and its degradation provides extra support for neighboring neurons during energy deficiency. Salvianolic acid B (SAB) exerts a neuroprotective effect on reperfusion insult after cerebrovascular occlusion, but the effect of SAB on astrocytic glycogen and its relationship with neuroprotection are not completely understood. Here, we knocked down astrocyte-specific glycogen phosphorylase (GP, the rate-limiting enzyme in glycogenolysis) in vitro and in vivo and investigated the changes in key enzymes in glycogen metabolism by performing immunoblotting in vitro and immunofluorescence in vivo. Neurobehavioral and morphological assessments were conducted to uncover the outcomes during brain reperfusion. SAB accelerated astrocytic glycogenolysis by upregulating GP activity but not GP expression after reperfusion. Suppression of astrocytic glycogenolysis weakened SAB-mediated neuroprotection against the reperfusion insult. In addition, activation of glycogenolysis by SAB contributed to the survival of astrocytes and surrounding neurons by increasing antioxidant levels in astrocytes. Our data reveal that astrocytic GP represents an important metabolic target in SAB-induced protection against brain damage after cerebrovascular recanalization.
Collapse
|
33
|
Nagano H, Ito S, Masuda T, Ohtsuki S. Effect of Insulin Receptor-Knockdown on the Expression Levels of Blood-Brain Barrier Functional Proteins in Human Brain Microvascular Endothelial Cells. Pharm Res 2021; 39:1561-1574. [PMID: 34811625 DOI: 10.1007/s11095-021-03131-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE The insulin receptor (INSR) mediates insulin signaling to modulate cellular functions. Although INSR is expressed at the blood-brain barrier (BBB), its role in the modulation of BBB function is poorly understood. Therefore, in this study, we aimed to analyze the effect of INSR knockdown on the expression levels of functional proteins at the BBB. METHODS We established the INSR-knockdown cell line (shINSR) using human cerebral microvascular endothelial cells (hCMEC/D3). The cellular proteome was analyzed using quantitative proteomics. RESULTS INSR mRNA and protein expressions were decreased in shINSR cells. The suppression of INSR-mediated signaling in shINSR cells was evaluated. The proteins involved in glycolysis and glycogenolysis were suppressed in shINSR cells. As amyloid-β peptide-related proteins, the expressions of presenilin-1 was increased, and those of the insulin-degrading enzyme and neprilysin were decreased. The expressions of BBB transporters, including the ABCB1/MDR1, ABCG2/BCRP, and SLCO2A1/OATP2A1 were significantly decreased by more than 50% in shINSR cells. The efflux activity of ABCB1/MDR1 was also suppressed. The expressions of the low-density lipoprotein receptor-related protein 1 were significantly increased, and those of the transferrin receptor were significantly decreased in shINSR cells. The expression of claudin-5 was also suppressed in shINSR cells. CONCLUSIONS The present study suggests that INSR-mediated signaling is involved in the regulation of functional protein expression at the BBB and contributes to the maintenance of BBB function. Changes in the expressions of amyloid-β peptide-related proteins may contribute to the development of cerebral amyloid angiopathy via the suppression of INSR-mediated signaling.
Collapse
Affiliation(s)
- Hinako Nagano
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| |
Collapse
|
34
|
Guo H, Fan Z, Wang S, Ma L, Wang J, Yu D, Zhang Z, Wu L, Peng Z, Liu W, Hou W, Cai Y. Astrocytic A1/A2 paradigm participates in glycogen mobilization mediated neuroprotection on reperfusion injury after ischemic stroke. J Neuroinflammation 2021; 18:230. [PMID: 34645472 PMCID: PMC8513339 DOI: 10.1186/s12974-021-02284-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Background Astrocytic glycogen works as an essential energy reserve for surrounding neurons and is reported to accumulate excessively during cerebral ischemia/reperfusion (I/R) injury. Our previous study found that accumulated glycogen mobilization exhibits a neuroprotective effect against I/R damage. In addition, ischemia could transform astrocytes into A1-like (toxic) and A2-like (protective) subtypes. However, the underlying mechanism behind accumulated glycogen mobilization-mediated neuroprotection in cerebral reperfusion injury and its relationship with the astrocytic A1/A2 paradigm is unknown. Methods Astrocytic glycogen phosphorylase, the rate-limiting enzyme in glycogen mobilization, was specifically overexpressed and knocked down in mice and in cultured astrocytes. The I/R injury was imitated using a middle cerebral artery occlusion/reperfusion model in mice and an oxygen–glucose deprivation/reoxygenation model in cultured cells. Alterations in A1-like and A2-like astrocytes and the expression of phosphorylated nuclear transcription factor-κB (NF-κB) and phosphorylated signal transducer and activator of transcription 3 (STAT3) were determined by RNA sequencing, immunofluorescence and immunoblotting. Metabolites, including glycogen, NADPH, glutathione and reactive oxygen species (ROS), were analyzed by biochemical analysis. Results Here, we observed that astrocytic glycogen mobilization inhibited A1-like astrocytes and enhanced A2-like astrocytes after reperfusion in an experimental ischemic stroke model in vivo and in vitro. In addition, glycogen mobilization could enhance the production of NADPH and glutathione by the pentose phosphate pathway (PPP) and reduce ROS levels during reperfusion. NF-κB inhibition and STAT3 activation caused by a decrease in ROS levels were responsible for glycogen mobilization-induced A1-like and A2-like astrocyte transformation after I/R. The astrocytic A1/A2 paradigm is closely correlated with glycogen mobilization-mediated neuroprotection in cerebral reperfusion injury. Conclusions Our data suggest that ROS-mediated NF-κB inhibition and STAT3 activation are the key pathways for glycogen mobilization-induced neuroprotection and provide a promising metabolic target for brain reperfusion injury in ischemic stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02284-y.
Collapse
Affiliation(s)
- Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ze Fan
- Department of Anesthesiology, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lina Ma
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jin Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Doutong Yu
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhen Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lin Wu
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhengwu Peng
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenming Liu
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yanhui Cai
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
35
|
Adiponectin Treatment Attenuates Cerebral Ischemia-Reperfusion Injury through HIF-1 α-Mediated Antioxidation in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5531048. [PMID: 34336097 PMCID: PMC8298180 DOI: 10.1155/2021/5531048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/01/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023]
Abstract
Adiponectin (ADPN) plays an important role in cerebral ischemia-reperfusion injury. Although previous studies have confirmed that ADPN pretreatment has a protective effect on ischemic stroke, the therapeutic effect of ADPN on ischemic stroke and the underlying mechanism are still unclear. In order to clarify these questions, focal transient cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in mice and ADPN was administered for three times at 6 h, 24 h, and 48 h after reperfusion. Meanwhile, a virus-delivered HIF-1α siRNA was used before ADPN administration. The infarct volume, neurological score, cellular apoptosis, and oxidative stress were assessed at 72 h after reperfusion. The long-term outcome of mice after stroke was recorded as well. The results indicated that ADPN treatment reduced the infarct volume (P = 0.032), neurological deficits (P = 0.047), cellular apoptosis (P = 0.041), and oxidative responses (P = 0.031) at 72 h after MCAO. Moreover, ADPN increased both the protein level and transcriptional activity of HIF-1α as evidenced by the transcription levels of VEGF (P = 0.046) and EPO (P = 0.043) at 72 h after MCAO. However, knockdown of HIF-1α partially reversed the antioxidant and treatment effect of ADPN after cerebral ischemia. In the observation of long-term outcome after ADPN treatment, it demonstrated that ADPN not only prevented the cerebral atrophy (P = 0.031) and the neurological function decline (P = 0.048), but also promoted angiogenesis (P = 0.028) after stroke. In conclusion, our findings suggest that ADPN is effective in treatment of ischemic stroke which could be attributed to the increased antioxidant capacity regulated by HIF-1α.
Collapse
|