1
|
Yang TH, Kang EYC, Lin PH, Yu BBC, Wang JHH, Chen V, Wang NK. Mitochondria in Retinal Ganglion Cells: Unraveling the Metabolic Nexus and Oxidative Stress. Int J Mol Sci 2024; 25:8626. [PMID: 39201313 PMCID: PMC11354650 DOI: 10.3390/ijms25168626] [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: 05/31/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
This review explored the role of mitochondria in retinal ganglion cells (RGCs), which are essential for visual processing. Mitochondrial dysfunction is a key factor in the pathogenesis of various vision-related disorders, including glaucoma, hereditary optic neuropathy, and age-related macular degeneration. This review highlighted the critical role of mitochondria in RGCs, which provide metabolic support, regulate cellular health, and respond to cellular stress while also producing reactive oxygen species (ROS) that can damage cellular components. Maintaining mitochondrial function is essential for meeting RGCs' high metabolic demands and ensuring redox homeostasis, which is crucial for their proper function and visual health. Oxidative stress, exacerbated by factors like elevated intraocular pressure and environmental factors, contributes to diseases such as glaucoma and age-related vision loss by triggering cellular damage pathways. Strategies targeting mitochondrial function or bolstering antioxidant defenses include mitochondrial-based therapies, gene therapies, and mitochondrial transplantation. These advances can offer potential strategies for addressing mitochondrial dysfunction in the retina, with implications that extend beyond ocular diseases.
Collapse
Affiliation(s)
- Tsai-Hsuan Yang
- Department of Education, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Eugene Yu-Chuan Kang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
| | - Pei-Hsuan Lin
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- National Taiwan University Hospital, Yunlin 640203, Taiwan
| | - Benjamin Ben-Chi Yu
- Fu Foundation School of Engineering & Applied Science, Columbia University, New York, NY 10027, USA;
| | - Jason Hung-Hsuan Wang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA
| | - Vincent Chen
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada
| | - Nan-Kai Wang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
2
|
Long Y, Zhao Z, Xie W, Shi J, Yang F, Zhu D, Jiang P, Tang Q, Ti Z, Jiang B, Yang X, Gao G, Qi W. Kallistatin leads to cognition impairment via downregulating glutamine synthetase. Pharmacol Res 2024; 202:107145. [PMID: 38492829 DOI: 10.1016/j.phrs.2024.107145] [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: 12/07/2023] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
In many neurodegenerative disorders, such as Alzheimer's disease (AD), glutamate-mediated neuronal excitotoxicity is considered the basis for cognitive impairment. The mRNA and protein expression of SERPINA4(Kallistatin) are higher in patients with AD. However, whether Kallistatin plays a regulatory role in glutamate-glutamine cycle homeostasis remains unclear. In this study, we identified impaired cognitive function in Kallistatin transgenic (KAL-TG) mice. Baseline glutamate levels were elevated and miniature excitatory postsynaptic current (mEPSC) frequency was increased in the hippocampus, suggesting the impairment of glutamate homeostasis in KAL-TG mice. Mechanistically, we demonstrated that Kallistatin promoted lysine acetylation and ubiquitination of glutamine synthetase (GS) and facilitated its degradation via the proteasome pathway, thereby downregulating GS. Fenofibrate improved cognitive memory in KAL-TG mice by downregulating serum Kallistatin. Collectively, our study findings provide insights the mechanism by which Kallistatin regulates cognitive impairment, and suggest the potential of fenofibrate to prevente and treat of AD patients with high levels of Kallistatin.
Collapse
Affiliation(s)
- Yanlan Long
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhen Zhao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wanting Xie
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinhui Shi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Fengyu Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dan Zhu
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Jiang
- Department of Clinical Medical Laboratory, Guangzhou First People Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qilong Tang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhou Ti
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bin Jiang
- Guangdong Province Key Laboratory of Brain Function and Disease, School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Xia Yang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Guoquan Gao
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong, China.
| | - Weiwei Qi
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products (Sun Yat-sen University), Guangzhou, China; Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
3
|
Blanco-Duque C, Chan D, Kahn MC, Murdock MH, Tsai LH. Audiovisual gamma stimulation for the treatment of neurodegeneration. J Intern Med 2024; 295:146-170. [PMID: 38115692 PMCID: PMC10842797 DOI: 10.1111/joim.13755] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Alzheimer's disease (AD) is the most common type of neurodegenerative disease and a health challenge with major social and economic consequences. In this review, we discuss the therapeutic potential of gamma stimulation in treating AD and delve into the possible mechanisms responsible for its positive effects. Recent studies reveal that it is feasible and safe to induce 40 Hz brain activity in AD patients through a range of 40 Hz multisensory and noninvasive electrical or magnetic stimulation methods. Although research into the clinical potential of these interventions is still in its nascent stages, these studies suggest that 40 Hz stimulation can yield beneficial effects on brain function, disease pathology, and cognitive function in individuals with AD. Specifically, we discuss studies involving 40 Hz light, auditory, and vibrotactile stimulation, as well as noninvasive techniques such as transcranial alternating current stimulation and transcranial magnetic stimulation. The precise mechanisms underpinning the beneficial effects of gamma stimulation in AD are not yet fully elucidated, but preclinical studies have provided relevant insights. We discuss preclinical evidence related to both neuronal and nonneuronal mechanisms that may be involved, touching upon the relevance of interneurons, neuropeptides, and specific synaptic mechanisms in translating gamma stimulation into widespread neuronal activity within the brain. We also explore the roles of microglia, astrocytes, and the vasculature in mediating the beneficial effects of gamma stimulation on brain function. Lastly, we examine upcoming clinical trials and contemplate the potential future applications of gamma stimulation in the management of neurodegenerative disorders.
Collapse
Affiliation(s)
- Cristina Blanco-Duque
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Diane Chan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Martin C Kahn
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mitchell H Murdock
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
Piotrowicz Z, Czuba M, Chalimoniuk M, Langfort J. The Impact of Acute Mild Normobaric Hypoxia and a Single Bout of Exercise to Volitional Exhaustion on Cognitive Performance in Endurance and Strength-Trained Athletes: The role of BDNF, EP-1, Catecholamines and Lactate. J Hum Kinet 2023; 87:77-93. [PMID: 37559758 PMCID: PMC10407317 DOI: 10.5114/jhk/168282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/05/2023] [Indexed: 08/11/2023] Open
Abstract
The aim of the study was to examine whether a single bout of exercise to volitional exhaustion, performed under moderate normobaric hypoxia (H), would affect psychomotor performance (PP) in differently trained athletes. For this purpose, ten strength-trained (S) athletes, ten endurance-trained (E) athletes and ten healthy men leading a sedentary lifestyle as a control (C) group performed voluntarily two graded exercise tests until volitional exhaustion (EVE) under normoxia (N) and H (FiO2 = 14.7%). We measured the peripheral level of the brain derived neurotrophic factor (BDNF), choice reaction time (CRT) and the number of correct reactions (NCR) as indices of PP. Psychomotor tests were performed at rest, immediately after the EVE and 3 minutes after the EVE. Venous blood samples were collected at rest, immediately after cessation of each EVE, and 1 h after each EVE. The results showed that the EVE significantly (p < 0.05) impaired CRT under N and H, and NCR under H only in the E group. The higher WRmax in the E compared to the S and C groups was associated with a significant (p < 0.005) increase in adrenaline (A) and noradrenaline (NA). There were no significant differences between conditions (N vs. H) in the BDNF at rest and after exercise. The EVE impaired cognitive function only in the E group; higher involvement of the sympathetic nervous system, A and NA may also play a role in this phenomenon. Therefore, it can be concluded that exposure to H did not have a negative impact on CRT or NCR. Moreover, BDNF did not improve cognitive function.
Collapse
Affiliation(s)
- Zofia Piotrowicz
- Institute of Sport Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Miłosz Czuba
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, Zielona Gora, Poland
| | - Małgorzata Chalimoniuk
- Department of Physical Education and Health in Biała Podlaska, Józef Piłsudski University of Physical Education in Warsaw, Biała Podlaska, Poland
| | - Józef Langfort
- Institute of Sport Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| |
Collapse
|
5
|
Crunfli F, Carregari VC, Veras FP, Silva LS, Nogueira MH, Antunes ASLM, Vendramini PH, Valença AGF, Brandão-Teles C, Zuccoli GDS, Reis-de-Oliveira G, Silva-Costa LC, Saia-Cereda VM, Smith BJ, Codo AC, de Souza GF, Muraro SP, Parise PL, Toledo-Teixeira DA, Santos de Castro ÍM, Melo BM, Almeida GM, Firmino EMS, Paiva IM, Silva BMS, Guimarães RM, Mendes ND, Ludwig RL, Ruiz GP, Knittel TL, Davanzo GG, Gerhardt JA, Rodrigues PB, Forato J, Amorim MR, Brunetti NS, Martini MC, Benatti MN, Batah SS, Siyuan L, João RB, Aventurato ÍK, Rabelo de Brito M, Mendes MJ, da Costa BA, Alvim MKM, da Silva Júnior JR, Damião LL, de Sousa IMP, da Rocha ED, Gonçalves SM, Lopes da Silva LH, Bettini V, Campos BM, Ludwig G, Tavares LA, Pontelli MC, Viana RMM, Martins RB, Vieira AS, Alves-Filho JC, Arruda E, Podolsky-Gondim GG, Santos MV, Neder L, Damasio A, Rehen S, Vinolo MAR, Munhoz CD, Louzada-Junior P, Oliveira RD, Cunha FQ, Nakaya HI, Mauad T, Duarte-Neto AN, Ferraz da Silva LF, Dolhnikoff M, Saldiva PHN, Farias AS, Cendes F, Moraes-Vieira PMM, Fabro AT, Sebollela A, Proença-Modena JL, Yasuda CL, Mori MA, Cunha TM, Martins-de-Souza D. Morphological, cellular, and molecular basis of brain infection in COVID-19 patients. Proc Natl Acad Sci U S A 2022. [DOI: 10.1073/pnas.2200960119 1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Although increasing evidence confirms neuropsychiatric manifestations associated mainly with severe COVID-19 infection, long-term neuropsychiatric dysfunction (recently characterized as part of “long COVID-19” syndrome) has been frequently observed after mild infection. We show the spectrum of cerebral impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, ranging from long-term alterations in mildly infected individuals (orbitofrontal cortical atrophy, neurocognitive impairment, excessive fatigue and anxiety symptoms) to severe acute damage confirmed in brain tissue samples extracted from the orbitofrontal region (via endonasal transethmoidal access) from individuals who died of COVID-19. In an independent cohort of 26 individuals who died of COVID-19, we used histopathological signs of brain damage as a guide for possible SARS-CoV-2 brain infection and found that among the 5 individuals who exhibited those signs, all of them had genetic material of the virus in the brain. Brain tissue samples from these five patients also exhibited foci of SARS-CoV-2 infection and replication, particularly in astrocytes. Supporting the hypothesis of astrocyte infection, neural stem cell–derived human astrocytes in vitro are susceptible to SARS-CoV-2 infection through a noncanonical mechanism that involves spike–NRP1 interaction. SARS-CoV-2–infected astrocytes manifested changes in energy metabolism and in key proteins and metabolites used to fuel neurons, as well as in the biogenesis of neurotransmitters. Moreover, human astrocyte infection elicits a secretory phenotype that reduces neuronal viability. Our data support the model in which SARS-CoV-2 reaches the brain, infects astrocytes, and consequently, leads to neuronal death or dysfunction. These deregulated processes could contribute to the structural and functional alterations seen in the brains of COVID-19 patients.
Collapse
Affiliation(s)
- Fernanda Crunfli
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Victor C. Carregari
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Flavio P. Veras
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Lucas S. Silva
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Mateus Henrique Nogueira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | | | - Pedro Henrique Vendramini
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | | | - Caroline Brandão-Teles
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Giuliana da Silva Zuccoli
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Guilherme Reis-de-Oliveira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Lícia C. Silva-Costa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Verônica Monteiro Saia-Cereda
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Bradley J. Smith
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Ana Campos Codo
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Gabriela F de Souza
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Stéfanie P. Muraro
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Pierina Lorencini Parise
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Daniel A. Toledo-Teixeira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | | | - Bruno Marcel Melo
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Glaucia M. Almeida
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | | | - Isadora Marques Paiva
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | | | - Rafaela Mano Guimarães
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Niele D. Mendes
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Raíssa L. Ludwig
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Gabriel P. Ruiz
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Thiago L. Knittel
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Gustavo G. Davanzo
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Jaqueline Aline Gerhardt
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Patrícia Brito Rodrigues
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Julia Forato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Mariene Ribeiro Amorim
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Natália S. Brunetti
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Matheus Cavalheiro Martini
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Maíra Nilson Benatti
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Sabrina S. Batah
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Li Siyuan
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Rafael B. João
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Ítalo K. Aventurato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Mariana Rabelo de Brito
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Maria J. Mendes
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Beatriz A. da Costa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Marina K. M. Alvim
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - José Roberto da Silva Júnior
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Lívia L. Damião
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Iêda Maria P. de Sousa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Elessandra D. da Rocha
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Solange M. Gonçalves
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Luiz H. Lopes da Silva
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Vanessa Bettini
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Brunno M. Campos
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Guilherme Ludwig
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Lucas Alves Tavares
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | | | | | - Ronaldo B. Martins
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Andre Schwambach Vieira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | | | - Eurico Arruda
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | | | - Marcelo Volpon Santos
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Luciano Neder
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - André Damasio
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Stevens Rehen
- D'Or Institute for Research and Education, 04502001, Brazil
- Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, 21941590, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | | | - Paulo Louzada-Junior
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Renê Donizeti Oliveira
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Fernando Q. Cunha
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | | | - Thais Mauad
- University of São Paulo, São Paulo, 05508-220, Brazil
| | | | | | | | | | - Alessandro S. Farias
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Fernando Cendes
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Pedro Manoel M. Moraes-Vieira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Alexandre T. Fabro
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Adriano Sebollela
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - José L. Proença-Modena
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Clarissa L. Yasuda
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Marcelo A. Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
| | - Thiago M. Cunha
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, 14049900, Brazil
| | - Daniel Martins-de-Souza
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, 13083862, Brazil
- D'Or Institute for Research and Education, 04502001, Brazil
| |
Collapse
|
6
|
Abstract
Although increasing evidence confirms neuropsychiatric manifestations associated mainly with severe COVID-19 infection, long-term neuropsychiatric dysfunction (recently characterized as part of "long COVID-19" syndrome) has been frequently observed after mild infection. We show the spectrum of cerebral impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, ranging from long-term alterations in mildly infected individuals (orbitofrontal cortical atrophy, neurocognitive impairment, excessive fatigue and anxiety symptoms) to severe acute damage confirmed in brain tissue samples extracted from the orbitofrontal region (via endonasal transethmoidal access) from individuals who died of COVID-19. In an independent cohort of 26 individuals who died of COVID-19, we used histopathological signs of brain damage as a guide for possible SARS-CoV-2 brain infection and found that among the 5 individuals who exhibited those signs, all of them had genetic material of the virus in the brain. Brain tissue samples from these five patients also exhibited foci of SARS-CoV-2 infection and replication, particularly in astrocytes. Supporting the hypothesis of astrocyte infection, neural stem cell-derived human astrocytes in vitro are susceptible to SARS-CoV-2 infection through a noncanonical mechanism that involves spike-NRP1 interaction. SARS-CoV-2-infected astrocytes manifested changes in energy metabolism and in key proteins and metabolites used to fuel neurons, as well as in the biogenesis of neurotransmitters. Moreover, human astrocyte infection elicits a secretory phenotype that reduces neuronal viability. Our data support the model in which SARS-CoV-2 reaches the brain, infects astrocytes, and consequently, leads to neuronal death or dysfunction. These deregulated processes could contribute to the structural and functional alterations seen in the brains of COVID-19 patients.
Collapse
|
7
|
de Oliveira Figueiredo EC, Bondiolotti BM, Laugeray A, Bezzi P. Synaptic Plasticity Dysfunctions in the Pathophysiology of 22q11 Deletion Syndrome: Is There a Role for Astrocytes? Int J Mol Sci 2022; 23:ijms23084412. [PMID: 35457231 PMCID: PMC9028090 DOI: 10.3390/ijms23084412] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 01/01/2023] Open
Abstract
The 22q11 deletion syndrome (DS) is the most common microdeletion syndrome in humans and gives a high probability of developing psychiatric disorders. Synaptic and neuronal malfunctions appear to be at the core of the symptoms presented by patients. In fact, it has long been suggested that the behavioural and cognitive impairments observed in 22q11DS are probably due to alterations in the mechanisms regulating synaptic function and plasticity. Often, synaptic changes are related to structural and functional changes observed in patients with cognitive dysfunctions, therefore suggesting that synaptic plasticity has a crucial role in the pathophysiology of the syndrome. Most interestingly, among the genes deleted in 22q11DS, six encode for mitochondrial proteins that, in mouse models, are highly expressed just after birth, when active synaptogenesis occurs, therefore indicating that mitochondrial processes are strictly related to synapse formation and maintenance of a correct synaptic signalling. Because correct synaptic functioning, not only requires correct neuronal function and metabolism, but also needs the active contribution of astrocytes, we summarize in this review recent studies showing the involvement of synaptic plasticity in the pathophysiology of 22q11DS and we discuss the relevance of mitochondria in these processes and the possible involvement of astrocytes.
Collapse
Affiliation(s)
| | - Bianca Maria Bondiolotti
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
| | - Anthony Laugeray
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland; (E.C.d.O.F.); (B.M.B.); (A.L.)
- Department of Pharmacology and Physiology, University of Rome Sapienza, 00185 Rome, Italy
- Correspondence: or
| |
Collapse
|
8
|
Brain Metabolic Alterations in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23073785. [PMID: 35409145 PMCID: PMC8998942 DOI: 10.3390/ijms23073785] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
The brain is one of the most energy-consuming organs in the body. Satisfying such energy demand requires compartmentalized, cell-specific metabolic processes, known to be complementary and intimately coupled. Thus, the brain relies on thoroughly orchestrated energy-obtaining agents, processes and molecular features, such as the neurovascular unit, the astrocyte-neuron metabolic coupling, and the cellular distribution of energy substrate transporters. Importantly, early features of the aging process are determined by the progressive perturbation of certain processes responsible for adequate brain energy supply, resulting in brain hypometabolism. These age-related brain energy alterations are further worsened during the prodromal stages of neurodegenerative diseases, namely Alzheimer's disease (AD), preceding the onset of clinical symptoms, and are anatomically and functionally associated with the loss of cognitive abilities. Here, we focus on concrete neuroenergetic features such as the brain's fueling by glucose and lactate, the transporters and vascular system guaranteeing its supply, and the metabolic interactions between astrocytes and neurons, and on its neurodegenerative-related disruption. We sought to review the principles underlying the metabolic dimension of healthy and AD brains, and suggest that the integration of these concepts in the preventive, diagnostic and treatment strategies for AD is key to improving the precision of these interventions.
Collapse
|
9
|
Mayorga-Weber G, Rivera FJ, Castro MA. Neuron-glia (mis)interactions in brain energy metabolism during aging. J Neurosci Res 2022; 100:835-854. [PMID: 35085408 DOI: 10.1002/jnr.25015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/08/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023]
Abstract
Life expectancy in humans is increasing, resulting in a growing aging population, that is accompanied by an increased disposition to develop cognitive deterioration. Hypometabolism is one of the multiple factors related to inefficient brain function during aging. This review emphasizes the metabolic interactions between glial cells (astrocytes, oligodendrocytes, and microglia) and neurons, particularly, during aging. Glial cells provide support and protection to neurons allowing adequate synaptic activity. We address metabolic coupling from the expression of transporters, availability of substrates, metabolic pathways, and mitochondrial activity. In aging, the main metabolic exchange machinery is altered with inefficient levels of nutrients and detrimental mitochondrial activity that results in high reactive oxygen species levels and reduced ATP production, generating a highly inflammatory environment that favors deregulated cell death. Here, we provide an overview of the glial-to-neuron mechanisms, from the molecular components to the cell types, emphasizing aging as the crucial risk factor for developing neurodegenerative/neuroinflammatory diseases.
Collapse
Affiliation(s)
- Gonzalo Mayorga-Weber
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco J Rivera
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Laboratory of Stem Cells and Neuroregeneration, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Janelia Research Campus, HHMI, Ashburn, VA, USA
| |
Collapse
|
10
|
Matthews DC, Mao X, Dowd K, Tsakanikas D, Jiang CS, Meuser C, Andrews RD, Lukic AS, Lee J, Hampilos N, Shafiian N, Sano M, David Mozley P, Fillit H, McEwen BS, Shungu DC, Pereira AC. Riluzole, a glutamate modulator, slows cerebral glucose metabolism decline in patients with Alzheimer's disease. Brain 2021; 144:3742-3755. [PMID: 34145880 PMCID: PMC8719848 DOI: 10.1093/brain/awab222] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/07/2021] [Accepted: 05/22/2021] [Indexed: 11/14/2022] Open
Abstract
Dysregulation of glutamatergic neural circuits has been implicated in a cycle of toxicity, believed among the neurobiological underpinning of Alzheimer's disease. Previously, we reported preclinical evidence that the glutamate modulator riluzole, which is FDA approved for the treatment of amyotrophic lateral sclerosis, has potential benefits on cognition, structural and molecular markers of ageing and Alzheimer's disease. The objective of this study was to evaluate in a pilot clinical trial, using neuroimaging biomarkers, the potential efficacy and safety of riluzole in patients with Alzheimer's disease as compared to placebo. A 6-month phase 2 double-blind, randomized, placebo-controlled study was conducted at two sites. Participants consisted of males and females, 50 to 95 years of age, with a clinical diagnosis of probable Alzheimer's disease, and Mini-Mental State Examination between 19 and 27. Ninety-four participants were screened, 50 participants who met inclusion criteria were randomly assigned to receive 50 mg riluzole (n = 26) or placebo (n = 24) twice a day. Twenty-two riluzole-treated and 20 placebo participants completed the study. Primary end points were baseline to 6 months changes in (i) cerebral glucose metabolism as measured with fluorodeoxyglucose-PET in prespecified regions of interest (hippocampus, posterior cingulate, precuneus, lateral temporal, inferior parietal, frontal); and (ii) changes in posterior cingulate levels of the neuronal viability marker N-acetylaspartate as measured with in vivo proton magnetic resonance spectroscopy. Secondary outcome measures were neuropsychological testing for correlation with neuroimaging biomarkers and in vivo measures of glutamate in posterior cingulate measured with magnetic resonance spectroscopy as a potential marker of target engagement. Measures of cerebral glucose metabolism, a well-established Alzheimer's disease biomarker and predictor of disease progression, declined significantly less in several prespecified regions of interest with the most robust effect in posterior cingulate, and effects in precuneus, lateral temporal, right hippocampus and frontal cortex in riluzole-treated participants in comparison to the placebo group. No group effect was found in measures of N-acetylaspartate levels. A positive correlation was observed between cognitive measures and regional cerebral glucose metabolism. A group × visit interaction was observed in glutamate levels in posterior cingulate, potentially suggesting engagement of glutamatergic system by riluzole. In vivo glutamate levels positively correlated with cognitive performance. These findings support our main primary hypothesis that cerebral glucose metabolism would be better preserved in the riluzole-treated group than in the placebo group and provide a rationale for more powered, longer duration studies of riluzole as a potential intervention for Alzheimer's disease.
Collapse
Affiliation(s)
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | | | | | | | - Caroline Meuser
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Ana S Lukic
- ADM Diagnostics Inc., Northbrook, IL 60062, USA
| | - Jihyun Lee
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nicholas Hampilos
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Neeva Shafiian
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mary Sano
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - P David Mozley
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Howard Fillit
- Alzheimer's Drug Discovery Foundation, New York, NY 10019, USA
| | | | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ana C Pereira
- The Rockefeller University, New York, NY 10065, USA
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
11
|
Nutrients, Cognitive Function, and Brain Aging: What We Have Learned from Dogs. Med Sci (Basel) 2021; 9:medsci9040072. [PMID: 34842769 PMCID: PMC8628994 DOI: 10.3390/medsci9040072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/29/2021] [Accepted: 11/13/2021] [Indexed: 12/29/2022] Open
Abstract
Due to a difference in genetics, environmental factors, and nutrition, just like in people, dogs age at different rates. Brain aging in people and dogs share similar morphological changes including irreversible cortical atrophy, cerebral amyloid angiopathy, and ventricular enlargement. Due to severe and irreversible brain atrophy, some aging dogs develop cognitive dysfunction syndrome (CDS), which is equivalent to dementia or Alzheimer’s disease (AD) in people. The risk factors and causes of CDS in dogs have not been fully investigated, but age, gender, oxidative stress, and deficiency of sex hormones appears to be associated with increased risk of accelerated brain aging and CDS in dogs. Both AD and CDS are incurable diseases at this moment, therefore more efforts should be focused on preventing or reducing brain atrophy and minimizing the risk of AD in people and CDS in dogs. Since brain atrophy leads to irreversible cognitive decline and dementia, an optimal nutritional solution should be able to not only enhance cognitive function during aging but also reduce irreversible brain atrophy. Up to now, only one nutritional intervention has demonstrated both cognition-enhancing benefits and atrophy-reducing benefits.
Collapse
|
12
|
Mulica P, Grünewald A, Pereira SL. Astrocyte-Neuron Metabolic Crosstalk in Neurodegeneration: A Mitochondrial Perspective. Front Endocrinol (Lausanne) 2021; 12:668517. [PMID: 34025580 PMCID: PMC8138625 DOI: 10.3389/fendo.2021.668517] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Converging evidence made clear that declining brain energetics contribute to aging and are implicated in the initiation and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's disease. Indeed, both pathologies involve instances of hypometabolism of glucose and oxygen in the brain causing mitochondrial dysfunction, energetic failure and oxidative stress. Importantly, recent evidence suggests that astrocytes, which play a key role in supporting neuronal function and metabolism, might contribute to the development of neurodegenerative diseases. Therefore, exploring how the neuro-supportive role of astrocytes may be impaired in the context of these disorders has great therapeutic potential. In the following, we will discuss some of the so far identified features underlining the astrocyte-neuron metabolic crosstalk. Thereby, special focus will be given to the role of mitochondria. Furthermore, we will report on recent advancements concerning iPSC-derived models used to unravel the metabolic contribution of astrocytes to neuronal demise. Finally, we discuss how mitochondrial dysfunction in astrocytes could contribute to inflammatory signaling in neurodegenerative diseases.
Collapse
Affiliation(s)
- Patrycja Mulica
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
13
|
Liu H, Prokosch V. Energy Metabolism in the Inner Retina in Health and Glaucoma. Int J Mol Sci 2021; 22:ijms22073689. [PMID: 33916246 PMCID: PMC8036449 DOI: 10.3390/ijms22073689] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma, the leading cause of irreversible blindness, is a heterogeneous group of diseases characterized by progressive loss of retinal ganglion cells (RGCs) and their axons and leads to visual loss and blindness. Risk factors for the onset and progression of glaucoma include systemic and ocular factors such as older age, lower ocular perfusion pressure, and intraocular pressure (IOP). Early signs of RGC damage comprise impairment of axonal transport, downregulation of specific genes and metabolic changes. The brain is often cited to be the highest energy-demanding tissue of the human body. The retina is estimated to have equally high demands. RGCs are particularly active in metabolism and vulnerable to energy insufficiency. Understanding the energy metabolism of the inner retina, especially of the RGCs, is pivotal for understanding glaucoma’s pathophysiology. Here we review the key contributors to the high energy demands in the retina and the distinguishing features of energy metabolism of the inner retina. The major features of glaucoma include progressive cell death of retinal ganglions and optic nerve damage. Therefore, this review focuses on the energetic budget of the retinal ganglion cells, optic nerve and the relevant cells that surround them.
Collapse
|
14
|
Fabbri R, Saracino E, Treossi E, Zamboni R, Palermo V, Benfenati V. Graphene glial-interfaces: challenges and perspectives. NANOSCALE 2021; 13:4390-4407. [PMID: 33599662 DOI: 10.1039/d0nr07824g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Graphene nanosheets are mechanically strong but flexible, electrically conductive and bio-compatible. Thus, due to these unique properties, they are being intensively studied as materials for the next generation of neural interfaces. Most of the literature focused on optimizing the interface between these materials and neurons. However, one of the most common causes of implant failure is the adverse inflammatory reaction of glial cells. These cells are not, as previously considered, just passive and supportive cells, but play a crucial role in the physiology and pathology of the nervous system, and in the interaction with implanted electrodes. Besides providing structural support to neurons, glia are responsible for the modulation of synaptic transmission and control of central and peripheral homeostasis. Accordingly, knowledge on the interaction between glia and biomaterials is essential to develop new implant-based therapies for the treatment of neurological disorders, such as epilepsy, brain tumours, and Alzheimer's and Parkinson's disease. This work provides an overview of the emerging literature on the interaction of graphene-based materials with glial cells, together with a complete description of the different types of glial cells and problems associated with them. We believe that this description will be important for researchers working in materials science and nanotechnology to develop new active materials to interface, measure and stimulate these cells.
Collapse
Affiliation(s)
- Roberta Fabbri
- Consiglio Nazionale delle Ricerche, Istituto per la Sintesi Organica e la Fotoreattività (CNR-ISOF), via Piero Gobetti 101, 40129 Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
15
|
Thoman ME, McKarns SC. Metabolomic Profiling in Neuromyelitis Optica Spectrum Disorder Biomarker Discovery. Metabolites 2020; 10:metabo10090374. [PMID: 32961928 PMCID: PMC7570337 DOI: 10.3390/metabo10090374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
There is no specific test for diagnosing neuromyelitis optica spectrum disorder (NMOSD), a disabling autoimmune disease of the central nervous system. Instead, diagnosis relies on ruling out other related disorders with overlapping clinical symptoms. An urgency for NMOSD biomarker discovery is underscored by adverse responses to treatment following misdiagnosis and poor prognosis following the delayed onset of treatment. Pathogenic autoantibiotics that target the water channel aquaporin-4 (AQP4) and myelin oligodendrocyte glycoprotein (MOG) contribute to NMOSD pathology. The importance of early diagnosis between AQP4-Ab+ NMOSD, MOG-Ab+ NMOSD, AQP4-Ab− MOG-Ab− NMOSD, and related disorders cannot be overemphasized. Here, we provide a comprehensive data collection and analysis of the currently known metabolomic perturbations and related proteomic outcomes of NMOSD. We highlight short chain fatty acids, lipoproteins, amino acids, and lactate as candidate diagnostic biomarkers. Although the application of metabolomic profiling to individual NMOSD patient care shows promise, more research is needed.
Collapse
Affiliation(s)
- Maxton E. Thoman
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Laboratory of TGF-β Biology, Epigenetics, and Cytokine Regulation, Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Susan C. McKarns
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Laboratory of TGF-β Biology, Epigenetics, and Cytokine Regulation, Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Department of Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Correspondence:
| |
Collapse
|
16
|
Marchetti B. Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease. Redox Biol 2020; 36:101664. [PMID: 32863224 PMCID: PMC7395594 DOI: 10.1016/j.redox.2020.101664] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation have long been recognized to contribute to Parkinson's disease (PD), a common movement disorder characterized by the selective loss of midbrain dopaminergic neurons (mDAn) of the substantia nigra pars compacta (SNpc). The causes and mechanisms still remain elusive, but a complex interplay between several genes and a number of interconnected environmental factors, are chiefly involved in mDAn demise, as they intersect the key cellular functions affected in PD, such as the inflammatory response, mitochondrial, lysosomal, proteosomal and autophagic functions. Nuclear factor erythroid 2 -like 2 (NFE2L2/Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling cascade, a vital pathway for mDAn neurogenesis and neuroprotection, emerge as critical intertwinned actors in mDAn physiopathology, as a decline of an Nrf2/Wnt/β-catenin prosurvival axis with age underlying PD mutations and a variety of noxious environmental exposures drive PD neurodegeneration. Unexpectedly, astrocytes, the so-called "star-shaped" cells, harbouring an arsenal of "beneficial" and "harmful" molecules represent the turning point in the physiopathological and therapeutical scenario of PD. Fascinatingly, "astrocyte's fil rouge" brings back to Nrf2/Wnt resilience, as boosting the Nrf2/Wnt resilience program rejuvenates astrocytes, in turn (i) mitigating nigrostriatal degeneration of aged mice, (ii) reactivating neural stem progenitor cell proliferation and neuron differentiation in the brain and (iii) promoting a beneficial immunomodulation via bidirectional communication with mDAns. Then, through resilience of Nrf2/Wnt/β-catenin anti-ageing, prosurvival and proregenerative molecular programs, it seems possible to boost the inherent endogenous self-repair mechanisms. Here, the cellular and molecular aspects as well as the therapeutical options for rejuvenating glia-neuron dialogue will be discussed together with major glial-derived mechanisms and therapies that will be fundamental to the identification of novel diagnostic tools and treatments for neurodegenerative diseases (NDs), to fight ageing and nigrostriatal DAergic degeneration and promote functional recovery.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology Section, Medical School, University of Catania, Via S. Sofia 65, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Neuropharmacology Section, Via Conte Ruggero 73, 94018, Troina, EN, Italy.
| |
Collapse
|
17
|
Sun Y, Wang Y, Chen ST, Chen YJ, Shen J, Yao WB, Gao XD, Chen S. Modulation of the Astrocyte-Neuron Lactate Shuttle System contributes to Neuroprotective action of Fibroblast Growth Factor 21. Am J Cancer Res 2020; 10:8430-8445. [PMID: 32724479 PMCID: PMC7381735 DOI: 10.7150/thno.44370] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
A viewpoint considering Alzheimer's disease (AD) as “type 3 diabetes” emphasizes the pivotal role of dysfunctional brain energy metabolism in AD. The hormone fibroblast growth factor 21 (FGF21) is a crucial regulator in energy metabolism; however, our understanding of the therapeutic potential and mechanisms underlying the effect of FGF21 on neurodegeneration of AD is far from complete. Methods: To further elucidate the effect of FGF21 on AD-related neurodegeneration, we used APP/PS1 transgenic mice to assess the effects of FGF21 on memory dysfunction, amyloid plaque pathology and pathological tau hyperphosphorylation. We also established an in vitro system to mimic astrocyte-neuron communication and an in vivo model of acute injury. Based on the in vivo and in vitro models, we analyzed the neuroprotective actions of FGF21 and pathways related to astrocyte-neuron communication and further focused on the astrocyte-neuron lactate shuttle system. Results: Here, we report that FGF21 can ameliorate Alzheimer-like neurodegeneration in APP/PS1 transgenic mice. We detected defects in the astrocyte-neuron lactate shuttle system in the in vivo and in vitro models of AD and identified FGF21 as a neuroprotective molecule that can rescue these deficits. Administration of FGF21 can alleviate memory dysfunction, amyloid plaque pathology and pathological tau hyperphosphorylation, and the function of FGF21 in neurodegeneration is mediated in part by monocarboxylate transporters (MCTs). In vivo evidence also suggests that FGF21 acts centrally in mice to exert its effects on neurodegeneration and energy metabolism via its regulation of MCTs. Conclusions: These results suggest that FGF21 alters metabolic parameters to mediate its neuroprotective functions. Modulation of the astrocyte-neuron lactate shuttle system can be one of the most efficient strategies for FGF21 in Alzheimer-like degeneration and contributes to improvements in brain metabolic defects and amyloid β-induced cytotoxicity. Our findings provide insights into the mechanisms underlying the effects of FGF21 on neurodegeneration and brain energy metabolism and suggest that FGF21 may have therapeutic value in the treatment of AD and other neurodegenerative diseases.
Collapse
|
18
|
Frazier HN, Ghoweri AO, Anderson KL, Lin RL, Popa GJ, Mendenhall MD, Reagan LP, Craven RJ, Thibault O. Elevating Insulin Signaling Using a Constitutively Active Insulin Receptor Increases Glucose Metabolism and Expression of GLUT3 in Hippocampal Neurons. Front Neurosci 2020; 14:668. [PMID: 32733189 PMCID: PMC7358706 DOI: 10.3389/fnins.2020.00668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/02/2020] [Indexed: 12/31/2022] Open
Abstract
Insulin signaling is an integral component of healthy brain function, with evidence of positive insulin-mediated alterations in synaptic integrity, cerebral blood flow, inflammation, and memory. However, the specific pathways targeted by this peptide remain unclear. Previously, our lab used a molecular approach to characterize the impact of insulin signaling on voltage-gated calcium channels and has also shown that acute insulin administration reduces calcium-induced calcium release in hippocampal neurons. Here, we explore the relationship between insulin receptor signaling and glucose metabolism using similar methods. Mixed, primary hippocampal cultures were infected with either a control lentivirus or one containing a constitutively active human insulin receptor (IRβ). 2-NBDG imaging was used to obtain indirect measures of glucose uptake and utilization. Other outcome measures include Western immunoblots of GLUT3 and GLUT4 on total membrane and cytosolic subcellular fractions. Glucose imaging data indicate that neurons expressing IRβ show significant elevations in uptake and rates of utilization compared to controls. As expected, astrocytes did not respond to the IRβ treatment. Quantification of Western immunoblots show that IRβ is associated with significant elevations in GLUT3 expression, particularly in the total membrane subcellular fraction, but did not alter GLUT4 expression in either fraction. Our work suggests that insulin plays a significant role in mediating neuronal glucose metabolism, potentially through an upregulation in the expression of GLUT3. This provides further evidence for a potential therapeutic mechanism underlying the beneficial impact of intranasal insulin in the clinic.
Collapse
Affiliation(s)
- Hilaree N Frazier
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Adam O Ghoweri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Katie L Anderson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Ruei-Lung Lin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Gabriel J Popa
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Michael D Mendenhall
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Rolf J Craven
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| |
Collapse
|
19
|
Shao S, Qin T, Qian W, Yue Y, Xiao Y, Li X, Zhang D, Wang Z, Ma Q, Lei J. Positive feedback in Cav-1-ROS signalling in PSCs mediates metabolic coupling between PSCs and tumour cells. J Cell Mol Med 2020; 24:9397-9408. [PMID: 32633891 PMCID: PMC7417714 DOI: 10.1111/jcmm.15596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022] Open
Abstract
Caveolin-1 (Cav-1) is the principal structural component of caveolae, and its dysregulation occurs in cancer. However, the role of Cav-1 in pancreatic cancer (PDAC) tumorigenesis and metabolism is largely unknown. In this study, we aimed to investigate the effect of pancreatic stellate cell (PSC) Cav-1 on PDAC metabolism and aggression. We found that Cav-1 is expressed at low levels in PDAC stroma and that the loss of stromal Cav-1 is associated with poor survival. In PSCs, knockdown of Cav-1 promoted the production of reactive oxygen species (ROS), while ROS production further reduced the expression of Cav-1. Positive feedback occurs in Cav-1-ROS signalling in PSCs, which promotes PDAC growth and induces stroma-tumour metabolic coupling in PDAC. In PSCs, positive feedback in Cav-1-ROS signalling induced a shift in energy metabolism to glycolysis, with up-regulated expression of glycolytic enzymes (hexokinase 2 (HK-2), 6-phosphofructokinase (PFKP) and pyruvate kinase isozyme type M2 (PKM2)) and transporter (Glut1) expression and down-regulated expression of oxidative phosphorylation (OXPHOS) enzymes (translocase of outer mitochondrial membrane 20 (TOMM20) and NAD(P)H dehydrogenase [quinone] 1 (NQO1)). These events resulted in high levels of glycolysis products such as lactate, which was secreted by up-regulated monocarboxylate transporter 4 (MCT4) in PSCs. Simultaneously, PDAC cells took up these glycolysis products (lactate) through up-regulated MCT1 to undergo OXPHOS, with down-regulated expression of glycolytic enzymes (HK-2, PFKP and PKM2) and up-regulated expression of OXPHOS enzymes (TOMM20 and NQO1). Interrupting the metabolic coupling between the stroma and tumour cells may be an effective method for tumour therapy.
Collapse
Affiliation(s)
- Shan Shao
- Department of Oncology, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yangyang Yue
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuqi Li
- Department of General Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dong Zhang
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianjun Lei
- Department of Hepatobiliary Surgery, First affiliated hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
20
|
Intertwined ROS and Metabolic Signaling at the Neuron-Astrocyte Interface. Neurochem Res 2020; 46:23-33. [PMID: 31989468 DOI: 10.1007/s11064-020-02965-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 12/23/2022]
Abstract
Metabolism and redox signalling share critical nodes in the nervous system. In the last years, a series of major findings have challenged the current vision on how neural reactive oxygen species (ROS) are produced and handled in the nervous system. Once regarded as deleterious by-products, ROS are now shown to be essential for a metabolic and redox crosstalk. In turn, this coupling defines neural viability and function to control behaviour or leading to neurodegeneration when compromised. Findings like a different assembly of mitochondrial respiratory supercomplexes in neurons and astrocytes stands behind a divergent production of ROS in either cell type, more prominent in astrocytes. ROS levels are however tightly controlled by an antioxidant machinery in astrocytes, assumed as more efficient than that of neurons, to regulate redox signalling. By exerting this control in ROS abundance, metabolic functions are finely tuned in both neural cells. Further, a higher engagement of mitochondrial respiration and oxidative function in neurons, underpinned by redox equivalents supplied from the pentose phosphate pathway and from glia, differs from the otherwise strong glycolytic capacity of astrocytes. Here, we recapitulate major findings on how ROS and metabolism differ between neural cells but merge to define reciprocal signalling pathways, ultimately defining neural function and fate.
Collapse
|
21
|
Wang Q, Hu Y, Wan J, Dong B, Sun J. Lactate: A Novel Signaling Molecule in Synaptic Plasticity and Drug Addiction. Bioessays 2019; 41:e1900008. [PMID: 31270822 DOI: 10.1002/bies.201900008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/20/2019] [Indexed: 12/21/2022]
Abstract
l-Lactate is emerging as a crucial regulatory nexus for energy metabolism in the brain and signaling transduction in synaptic plasticity, memory processes, and drug addiction instead of being merely a waste by-product of anaerobic glycolysis. In this review, the role of lactate in various memory processes, synapse plasticity and drug addiction on the basis of recent studies is summarized and discussed. To this end, three main parts are presented: first, lactate as an energy substrate in energy metabolism of the brain is described; second, lactate as a novel signaling molecule in synaptic plasticity, neural circuits, memory, and drug addiction is described; and third, in light of the above descriptions, it is plausible to speculate that lactate is predominantly a signaling molecule in specific memory processes and partly acts as an energy substrate. The future perspective in lactate signaling involving microglia and associated precise signaling pathways in the brain is highlighted.
Collapse
Affiliation(s)
- Qiuting Wang
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ying Hu
- Department of Paediatrics, Jinan Zhangqiu District Hospital of TCM, Jinan, Shandong, 250200, China
| | - Jiale Wan
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China.,Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China
| | - Jinhao Sun
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Anatomy, School of Basic Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
22
|
Bourdon AK, Spano GM, Marshall W, Bellesi M, Tononi G, Serra PA, Baghdoyan HA, Lydic R, Campagna SR, Cirelli C. Metabolomic analysis of mouse prefrontal cortex reveals upregulated analytes during wakefulness compared to sleep. Sci Rep 2018; 8:11225. [PMID: 30046159 PMCID: PMC6060152 DOI: 10.1038/s41598-018-29511-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
By identifying endogenous molecules in brain extracellular fluid metabolomics can provide insight into the regulatory mechanisms and functions of sleep. Here we studied how the cortical metabolome changes during sleep, sleep deprivation and spontaneous wakefulness. Mice were implanted with electrodes for chronic sleep/wake recording and with microdialysis probes targeting prefrontal and primary motor cortex. Metabolites were measured using ultra performance liquid chromatography-high resolution mass spectrometry. Sleep/wake changes in metabolites were evaluated using partial least squares discriminant analysis, linear mixed effects model analysis of variance, and machine-learning algorithms. More than 30 known metabolites were reliably detected in most samples. When used by a logistic regression classifier, the profile of these metabolites across sleep, spontaneous wake, and enforced wake was sufficient to assign mice to their correct experimental group (pair-wise) in 80-100% of cases. Eleven of these metabolites showed significantly higher levels in awake than in sleeping mice. Some changes extend previous findings (glutamate, homovanillic acid, lactate, pyruvate, tryptophan, uridine), while others are novel (D-gluconate, N-acetyl-beta-alanine, N-acetylglutamine, orotate, succinate/methylmalonate). The upregulation of the de novo pyrimidine pathway, gluconate shunt and aerobic glycolysis may reflect a wake-dependent need to promote the synthesis of many essential components, from nucleic acids to synaptic membranes.
Collapse
Affiliation(s)
- Allen K Bourdon
- Department of Chemistry, University of Tennessee, Knoxville, TN, United States
| | - Giovanna Maria Spano
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - William Marshall
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - Michele Bellesi
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - Pier Andrea Serra
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Helen A Baghdoyan
- Department of Anesthesiology and Psychology, University of Tennessee, Knoxville, TN, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Ralph Lydic
- Department of Anesthesiology and Psychology, University of Tennessee, Knoxville, TN, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN, United States. .,Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN, United States.
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States.
| |
Collapse
|
23
|
Madji Hounoum B, Blasco H, Coque E, Vourc'h P, Emond P, Corcia P, Andres CR, Raoul C, Mavel S. The Metabolic Disturbances of Motoneurons Exposed to Glutamate. Mol Neurobiol 2018; 55:7669-7676. [PMID: 29435916 DOI: 10.1007/s12035-018-0945-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
Glutamate-induced excitotoxicity is considered as one of the major pathophysiological factors of motoneuron death in amyotrophic lateral sclerosis and other motoneuron diseases. In order to expand our knowledge on mechanisms of glutamate-induced excitotoxicity, the present study proposes to determine the metabolic consequences of glutamate and astrocytes in primary enriched motoneuron culture. Using liquid chromatography coupled to high-resolution mass spectrometry (LC-HRMS), we showed that the presence of astrocytes and glutamate profoundly modified the metabolic profile of motoneurons. Our study highlights for the first time that crosstalk between astrocytes and enriched motoneuron culture induced alterations in phenylalanine, tryptophan, purine, arginine, proline, aspartate, and glutamate metabolism in motoneurons. We observed that astrocytes modulate the sensitivity of motoneurons to glutamate, since metabolites altered by glutamate in motoneurons cultured alone were different (except 5-hydroxylysine) from those altered in co-cultured motoneurons. Our findings provide new insight into the metabolic alterations associated to astrocytes and glutamate in motoneurons and provide opportunities to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Blandine Madji Hounoum
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France
| | - Hélène Blasco
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France.,Laboratoire de Biochimie et de Biologie Moléculaire, Hôpital Bretonneau, CHRU de Tours, Tours, France
| | - Emmanuelle Coque
- The Institute for Neurosciences of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France
| | - Patrick Vourc'h
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France.,Laboratoire de Biochimie et de Biologie Moléculaire, Hôpital Bretonneau, CHRU de Tours, Tours, France
| | - Patrick Emond
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France
| | - Philippe Corcia
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France.,Centre SLA, Service de Neurologie, CHRU de Tours, Tours, France
| | - Christian R Andres
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France.,Laboratoire de Biochimie et de Biologie Moléculaire, Hôpital Bretonneau, CHRU de Tours, Tours, France
| | - Cédric Raoul
- The Institute for Neurosciences of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France
| | - Sylvie Mavel
- INSERM U930 "Imagerie et Cerveau," CHRU de Tours, Université François-Rabelais, Tours, France. .,UFR Pharmacie, INSERM U930, 31 av Monge, 37200, Tours, France.
| |
Collapse
|
24
|
Disrupted Neuroglial Metabolic Coupling after Peripheral Surgery. J Neurosci 2017; 38:452-464. [PMID: 29175959 DOI: 10.1523/jneurosci.1797-17.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 11/07/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023] Open
Abstract
Immune-related events in the periphery can remotely affect brain function, contributing to neurodegenerative processes and cognitive decline. In mice, peripheral surgery induces a systemic inflammatory response associated with changes in hippocampal synaptic plasticity and transient cognitive decline, however, the underlying mechanisms remain unknown. Here we investigated the effect of peripheral surgery on neuronal-glial function within hippocampal neuronal circuits of relevance to cognitive processing in male mice at 6, 24, and 72 h postsurgery. At 6 h we detect the proinflammatory cytokine IL-6 in the hippocampus, followed up by alterations in the mRNA and protein expression of astrocytic and neuronal proteins necessary for optimal energy supply to the brain and for the reuptake and recycling of glutamate in the synapse. Similarly, at 24 h postsurgery the mRNA expression of structural proteins (GFAP and AQP4) was compromised. At this time point, functional analysis in astrocytes revealed a decrease in resting calcium signaling. Examination of neuronal activity by whole-cell patch-clamp shows elevated levels of glutamatergic transmission and changes in AMPA receptor subunit composition at 72 h postsurgery. Finally, lactate, an essential energy substrate produced by astrocytes and critical for memory formation, decreases at 6 and 72 h after surgery. Based on temporal parallels with our previous studies, we propose that the previously reported cognitive decline observed at 72 h postsurgery in mice might be the consequence of temporal hippocampal metabolic, structural, and functional changes in astrocytes that lead to a disruption of the neuroglial metabolic coupling and consequently to a neuronal dysfunction.SIGNIFICANCE STATEMENT A growing body of evidence suggests that surgical trauma launches a systemic inflammatory response that reaches the brain and associates with immune activation and cognitive decline. Understanding the mechanisms by which immune-related events in the periphery can influence brain processes is essential for the development of therapies to prevent or treat postoperative cognitive dysfunction and other forms of cognitive decline related to immune-to-brain communication, such as Alzheimer's and Parkinson's diseases. Here we describe the temporal orchestration of a series of metabolic, structural, and functional changes after aseptic trauma in mice related to astrocytes and later in neurons that emphasize the role of astrocytes as key intermediaries between peripheral immune events, neuronal processing, and potentially cognition.
Collapse
|
25
|
Repetitive transcranial magnetic stimulation inhibits Sirt1/MAO-A signaling in the prefrontal cortex in a rat model of depression and cortex-derived astrocytes. Mol Cell Biochem 2017; 442:59-72. [PMID: 28948423 DOI: 10.1007/s11010-017-3193-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/09/2017] [Indexed: 02/06/2023]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a useful monotherapy for depression or adjunctive therapy for resistant depression. However, the anti-depressive effects of different parameters and the underlying mechanisms remain unclear. Here, we aimed to assess the effect of rTMS with different parameters (1/5/10 Hz, 0.84/1.26 T) on the depressive-like behaviors, 5-hydroxytryptamine (5-HT), 5-HIAA (5-hydroxyindoleacetic acid) and DA and NE levels, and monoamine oxidase A (MAO-A) activity in chronic unpredictable stress-treated rats, along with the expression of sirtuin 1 (Sirt1) and MAO-A in the prefrontal cortex (PFC) and cortex-derived astrocytes from new-born rats. Moreover, the depressive-like behaviors were monitored following the transcranial injection of the Sirt1 inhibitor EX527 (1 mM) daily for 1 week. We found that rTMS treatment (5/10 Hz, 0.84/1.26 T) ameliorated depressive-like behaviors, increased 5-HT, DA and NE levels, decreased the 5-HIAA level and Sirt1 and MAO-A expression, and reduced MAO-A activity in the PFC. The depressive-like behaviors were also ameliorated after the transcranial injection of EX527. Importantly, rTMS (5/10 Hz, 0.84/1.26 T) inhibited Sirt1 and MAO-A expressions in astrocytes and Sirt1 knockdown with short hairpin RNA decreased MAO-A expression in astrocytes. These results suggest that the inhibition of Sirt1/MAO-A expression in astrocytes in the PFC may contribute to the different anti-depressive effects of rTMS with different parameters, and may also provide a novel insight into the mechanisms underlying major depressive disorder.
Collapse
|
26
|
Anandhan A, Jacome MS, Lei S, Hernandez-Franco P, Pappa A, Panayiotidis MI, Powers R, Franco R. Metabolic Dysfunction in Parkinson's Disease: Bioenergetics, Redox Homeostasis and Central Carbon Metabolism. Brain Res Bull 2017; 133:12-30. [PMID: 28341600 PMCID: PMC5555796 DOI: 10.1016/j.brainresbull.2017.03.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 03/19/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022]
Abstract
The loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of protein inclusions (Lewy bodies) are the pathological hallmarks of Parkinson's disease (PD). PD is triggered by genetic alterations, environmental/occupational exposures and aging. However, the exact molecular mechanisms linking these PD risk factors to neuronal dysfunction are still unclear. Alterations in redox homeostasis and bioenergetics (energy failure) are thought to be central components of neurodegeneration that contribute to the impairment of important homeostatic processes in dopaminergic cells such as protein quality control mechanisms, neurotransmitter release/metabolism, axonal transport of vesicles and cell survival. Importantly, both bioenergetics and redox homeostasis are coupled to neuro-glial central carbon metabolism. We and others have recently established a link between the alterations in central carbon metabolism induced by PD risk factors, redox homeostasis and bioenergetics and their contribution to the survival/death of dopaminergic cells. In this review, we focus on the link between metabolic dysfunction, energy failure and redox imbalance in PD, making an emphasis in the contribution of central carbon (glucose) metabolism. The evidence summarized here strongly supports the consideration of PD as a disorder of cell metabolism.
Collapse
Affiliation(s)
- Annadurai Anandhan
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States; Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Maria S Jacome
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States
| | - Shulei Lei
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Pablo Hernandez-Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States; Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100 Alexandroupolis, Greece
| | | | - Robert Powers
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States; Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68503, United States
| | - Rodrigo Franco
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68516, United States; Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68503, United States.
| |
Collapse
|
27
|
Abstract
OBJECTIVE The exact pathophysiology of bipolar disorder (BD) is not yet fully understood, and there are many questions in this area which should be answered. This review aims to discuss the roles of glial cells in the pathophysiology of BD and their contribution to the mechanism of action of mood-stabilising drugs. METHODS We critically reviewed the most recent advances regarding glial cell roles in the pathophysiology and treatment of BD and the neuroprotective and neurotrophic effects of these cells. RESULTS Postmortem studies revealed a decrease in the glial cell number or density in the specific layers of prefrontal and anterior cingulate cortex in the patients with BD, whereas there was no difference in other brain regions, such as entorhinal cortex, amygdala and hippocampus. Astrocytes and oligodendrocytes were the most important glial types that were responsible for the glial reduction, but microglia activation rather than loss may be implicated in BD. The decreased number or density of glial cells may contribute to the pathological changes observed in neurons in the patients with BD. Alteration of specific neurotrophic factors such as glial cell line-derived neurotrophic factor and S100B may be an important feature of BD. Glial cells mediate the therapeutic effects of mood-stabilising agents in the treatment of BD. CONCLUSION Recent studies provide important evidence on the impairment of glial cells in the pathophysiology and treatment of BD. However, future controlled studies are necessary to elucidate different aspects of glial cells contribution to BD, and the mechanism of action of mood-stabilising drugs.
Collapse
|
28
|
Schnepp PM, Lee DD, Guldner IH, O'Tighearnaigh TK, Howe EN, Palakurthi B, Eckert KE, Toni TA, Ashfeld BL, Zhang S. GAD1 Upregulation Programs Aggressive Features of Cancer Cell Metabolism in the Brain Metastatic Microenvironment. Cancer Res 2017; 77:2844-2856. [PMID: 28400476 DOI: 10.1158/0008-5472.can-16-2289] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/19/2016] [Accepted: 04/03/2017] [Indexed: 11/16/2022]
Abstract
The impact of altered amino acid metabolism on cancer progression is not fully understood. We hypothesized that a metabolic transcriptome shift during metastatic evolution is crucial for brain metastasis. Here, we report a powerful impact in this setting caused by epigenetic upregulation of glutamate decarboxylase 1 (GAD1), a regulator of the GABA neurotransmitter metabolic pathway. In cell-based culture and brain metastasis models, we found that downregulation of the DNA methyltransferase DNMT1 induced by the brain microenvironment-derived clusterin resulted in decreased GAD1 promoter methylation and subsequent upregulation of GAD1 expression in brain metastatic tumor cells. In a system to dynamically visualize cellular metabolic responses mediated by GAD1, we monitored the cytosolic NADH:NAD+ equilibrium in tumor cells. Reducing GAD1 in metastatic cells by primary glia cell coculture abolished the capacity of metastatic cells to utilize extracellular glutamine, leading to cytosolic accumulation of NADH and increased oxidative status. Similarly, genetic or pharmacologic disruption of the GABA metabolic pathway decreased the incidence of brain metastasis in vivo Taken together, our results show how epigenetic changes in GAD1 expression alter local glutamate metabolism in the brain metastatic microenvironment, contributing to a metabolic adaption that facilitates metastasis outgrowth in that setting. Cancer Res; 77(11); 2844-56. ©2017 AACR.
Collapse
Affiliation(s)
- Patricia M Schnepp
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Dennis D Lee
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana
| | - Ian H Guldner
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Treasa K O'Tighearnaigh
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana
| | - Erin N Howe
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Bhavana Palakurthi
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana
| | - Kaitlyn E Eckert
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Tiffany A Toni
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Brandon L Ashfeld
- Mike and Josie Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana.,Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Siyuan Zhang
- Department of Biological Sciences, College of Science, University of Notre Dame, Notre Dame, Indiana. .,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana.,Indiana University Melvin & Bren Simon Cancer Center, Indianapolis, Indiana
| |
Collapse
|
29
|
Winkler U, Seim P, Enzbrenner Y, Köhler S, Sicker M, Hirrlinger J. Activity-dependent modulation of intracellular ATP in cultured cortical astrocytes. J Neurosci Res 2017; 95:2172-2181. [PMID: 28151554 DOI: 10.1002/jnr.24020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 01/21/2023]
Abstract
Brain function is absolutely dependent on an appropriate supply of energy. A shortfall in supply-as occurs, for instance, following stroke-can lead rapidly to irreversible damage to this vital organ. While the consequences of pathophysiological energy depletion have been well documented, much less is known about the physiological energy dynamics of brain cells, although changes in the intracellular concentration of adenosine triphosphate (ATP), the major energy carrier of cells, have been postulated to contribute to cellular signaling. To address this issue more closely, we have investigated intracellular ATP in cultured primary cortical astrocytes by time-lapse microscopy using a genetically encoded fluorescent sensor for ATP. The cytosolic ATP sensor signal decreased after application of the neurotransmitter glutamate in a manner dependent on both glutamate concentration and glutamate transporter activity, but independent of glutamate receptors. The application of dopamine did not affect ATP levels within astrocytes. These results confirm that intracellular ATP levels in astrocytes do indeed respond to changes in physiological activity and pave the way for further studies addressing factors that affect regulation of ATP. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ulrike Winkler
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Pauline Seim
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Yvonne Enzbrenner
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Susanne Köhler
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Marit Sicker
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Department of Neurogenetics, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
30
|
Mason S. Lactate Shuttles in Neuroenergetics-Homeostasis, Allostasis and Beyond. Front Neurosci 2017; 11:43. [PMID: 28210209 PMCID: PMC5288365 DOI: 10.3389/fnins.2017.00043] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/20/2017] [Indexed: 12/19/2022] Open
Abstract
Understanding brain energy metabolism—neuroenergetics—is becoming increasingly important as it can be identified repeatedly as the source of neurological perturbations. Within the scientific community we are seeing a shift in paradigms from the traditional neurocentric view to that of a more dynamic, integrated one where astrocytes are no longer considered as being just supportive, and activated microglia have a profound influence. Lactate is emerging as the “good guy,” contrasting its classical “bad guy” position in the now superseded medical literature. This review begins with the evolution of the concept of “lactate shuttles”; goes on to the recent shift in ideas regarding normal neuroenergetics (homeostasis)—specifically, the astrocyte–neuron lactate shuttle; and progresses to covering the metabolic implications whereby homeostasis is lost—a state of allostasis, and the function of microglia. The role of lactate, as a substrate and shuttle, is reviewed in light of allostatic stress, and beyond—in an acute state of allostatic stress in terms of physical brain trauma, and reflected upon with respect to persistent stress as allostatic overload—neurodegenerative diseases. Finally, the recently proposed astrocyte–microglia lactate shuttle is discussed in terms of chronic neuroinflammatory infectious diseases, using tuberculous meningitis as an example. The novelty extended by this review is that the directionality of lactate, as shuttles in the brain, in neuropathophysiological states is emerging as crucial in neuroenergetics.
Collapse
Affiliation(s)
- Shayne Mason
- Centre for Human Metabolomics, North-West University Potchefstroom, South Africa
| |
Collapse
|
31
|
Kadłubowska J, Malaguarnera L, Wąż P, Zorena K. Neurodegeneration and Neuroinflammation in Diabetic Retinopathy: Potential Approaches to Delay Neuronal Loss. Curr Neuropharmacol 2017; 14:831-839. [PMID: 27306035 PMCID: PMC5333588 DOI: 10.2174/1570159x14666160614095559] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 08/11/2015] [Accepted: 01/01/1970] [Indexed: 02/06/2023] Open
Abstract
In spite of the extensive research the complex pathogenesis of diabetic retinopathy (DR) has not been fully elucidated. For many years it has been thought that diabetic retinopathy manifests only with microangiopathic lesions, which are totally responsible for the loss of vision in diabetic patients. In view of the current knowledge on the microangiopathic changes in the fundus of the eye, diabetic retinopathy is perceived as a neurodegenerative disease. Several clinical tools are available to detect neuronal dysfunction at early stages of diabetes. Many functional changes in the retina can be identified before vascular pathology develops, suggesting that they result from a direct effect of diabetes on the neural retina. In the course of diabetes there is a chronic loss of retinal neurons due to increased frequency of apoptosis. The neuronal apoptosis begins very early in the course of diabetes. This observation has led to suggestions that precautions against DR should be implemented immediately after diabetes is diagnosed. Neurodegeneration cannot be reversed; therefore treatments preventing neuronal cell loss in the retina need to be developed to protect diabetic patients. This review is an attempt to summarize what is currently known about the mechanisms of neuronal apoptosis in the context of diabetic retinopathy and vascular degeneration as well as about potential treatments of DR
Collapse
Affiliation(s)
| | | | | | - Katarzyna Zorena
- Department of Immunobiology and Environment Microbiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| |
Collapse
|
32
|
|
33
|
Xu H, Zhang H, Zhang J, Huang Q, Shen Z, Wu R. Evaluation of neuron-glia integrity by in vivo proton magnetic resonance spectroscopy: Implications for psychiatric disorders. Neurosci Biobehav Rev 2016; 71:563-577. [PMID: 27702600 DOI: 10.1016/j.neubiorev.2016.09.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/18/2016] [Accepted: 09/26/2016] [Indexed: 02/05/2023]
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) has been widely applied in human studies. There is now a large literature describing findings of brain MRS studies with mental disorder patients including schizophrenia, bipolar disorder, major depressive disorder, and anxiety disorders. However, the findings are mixed and cannot be reconciled by any of the existing interpretations. Here we proposed the new theory of neuron-glia integrity to explain the findings of brain 1H-MRS stuies. It proposed the neurochemical correlates of neuron-astrocyte integrity and axon-myelin integrity on the basis of update of neurobiological knowledge about neuron-glia communication and of experimental MRS evidence for impairments in neuron-glia integrity from the authors and the other investigators. Following the neuron-glia integrity theories, this review collected evidence showing that glutamate/glutamine change is a good marker for impaired neuron-astrocyte integrity and that changes in N-acetylaspartate and lipid precursors reflect impaired myelination. Moreover, this new theory enables us to explain the differences between MRS findings in neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Haiyun Xu
- The Mental Health Center, Shantou University Medical College, China.
| | - Handi Zhang
- The Mental Health Center, Shantou University Medical College, China
| | - Jie Zhang
- The Mental Health Center, Shantou University Medical College, China
| | - Qingjun Huang
- The Mental Health Center, Shantou University Medical College, China
| | - Zhiwei Shen
- The Department of Radiology, the second affiliated hospital, Shantou University Medical College, China
| | - Renhua Wu
- The Department of Radiology, the second affiliated hospital, Shantou University Medical College, China
| |
Collapse
|
34
|
Patin F, Corcia P, Vourc’h P, Nadal-Desbarats L, Baranek T, Goossens JF, Marouillat S, Dessein AF, Descat A, Madji Hounoum B, Bruno C, Leman S, Andres CR, Blasco H. Omics to Explore Amyotrophic Lateral Sclerosis Evolution: the Central Role of Arginine and Proline Metabolism. Mol Neurobiol 2016; 54:5361-5374. [DOI: 10.1007/s12035-016-0078-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/23/2016] [Indexed: 12/13/2022]
|
35
|
The Protein Tyrosine Kinase Inhibitor Tyrphostin 23 Strongly Accelerates Glycolytic Lactate Production in Cultured Primary Astrocytes. Neurochem Res 2016; 41:2607-2618. [PMID: 27278759 DOI: 10.1007/s11064-016-1972-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/29/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
Tyrphostin 23 (T23) is a well-known inhibitor of protein tyrosine kinases. To investigate potential acute effects of T23 on the viability and the glucose metabolism of brain cells, we exposed cultured primary rat astrocytes to T23 for up to 4 h. While the viability and the morphology of the cultured astrocytes were not acutely affected by the presence of T23 in concentrations of up to 300 µM, this compound caused a rapid, time- and concentration-dependent increase in glucose consumption and lactate release. Maximal effects on glycolytic flux were found for incubations with 100 µM T23 for 2 h which doubled both glucose consumption and lactate production. The stimulation of glycolytic flux by T23 was reversible, completely abolished upon removal of the compound and not found in presence of other known inhibitors of endocytosis. Structurally related compounds such as tyrphostin 25 and catechol or modulators of AMP kinase activity did neither affect the basal nor the T23-stimulated lactate production by astrocytes. In contrast, the presence of the phosphatase inhibitor vanadate completely abolished the stimulation by T23 of astrocytic lactate production in a concentration-dependent manner. These data suggest that T23-sensitive phosphorylation/dephosphorylation events are involved in the regulation of astrocytic glycolysis.
Collapse
|
36
|
Ze X, Su M, Zhao X, Jiang H, Hong J, Yu X, Liu D, Xu B, Sheng L, Zhou Q, Zhou J, Cui J, Li K, Wang L, Ze Y, Hong F. TiO2 nanoparticle-induced neurotoxicity may be involved in dysfunction of glutamate metabolism and its receptor expression in mice. ENVIRONMENTAL TOXICOLOGY 2016; 31:655-662. [PMID: 25411160 DOI: 10.1002/tox.22077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 11/02/2014] [Accepted: 11/03/2014] [Indexed: 06/04/2023]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) have been used in environmental management, food, medicine, and industry. But TiO2 NPs have been demonstrated to cross the blood-brain barrier and store up in the brain organization, leading to glutamate-mediated neurotoxicity. However, the neurotoxicity in the brain is not well understood. In this study, mice were exposed to 1.25, 2.5, or 5 mg/kg body weight TiO2 NPs for 9 months, and the glutamate-glutamine cyclic pathway and expressions of glutamate receptors associated with the hippocampal neurotoxicity were investigated. Our findings showed elevations of glutamate release and phosphate-activated glutaminase activity, and reductions in glutamine and glutamine synthetase in the hippocampus following exposure to TiO2 NPs. Furthermore, TiO2 NPs significantly inhibited the expression of N-methyl-d-aspartate receptor subunits (including NR1, NR2A, and NR2B) and metabotropic glutamate receptor 2 in mouse hippocampus. These findings suggest that the imbalance of glutamate metabolism triggered inhibitions of glutamate receptor expression in the TiO2 NP-exposed hippocampus. © 2014 Wiley Periodicals, Inc. Environ Toxicol 31: 655-662, 2016.
Collapse
Affiliation(s)
- Xiao Ze
- Medical College of Soochow University, Suzhou, 215123, China
| | - Mingyu Su
- Suzhou Environmental Monitor Center, Suzhou, 215004, China
| | - Xiaoyang Zhao
- Medical College of Soochow University, Suzhou, 215123, China
| | - Hao Jiang
- Medical College of Soochow University, Suzhou, 215123, China
| | - Jie Hong
- Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaohong Yu
- Medical College of Soochow University, Suzhou, 215123, China
| | - Dong Liu
- Medical College of Soochow University, Suzhou, 215123, China
| | - Bingqing Xu
- Medical College of Soochow University, Suzhou, 215123, China
| | - Lei Sheng
- Medical College of Soochow University, Suzhou, 215123, China
| | - Qiuping Zhou
- Medical College of Soochow University, Suzhou, 215123, China
| | - Junling Zhou
- Medical College of Soochow University, Suzhou, 215123, China
| | - Jingwen Cui
- Medical College of Soochow University, Suzhou, 215123, China
| | - Kai Li
- Medical College of Soochow University, Suzhou, 215123, China
| | - Ling Wang
- Library of Soochow University, Suzhou, 215021, China
| | - Yuguan Ze
- Medical College of Soochow University, Suzhou, 215123, China
| | - Fashui Hong
- Medical College of Soochow University, Suzhou, 215123, China
| |
Collapse
|
37
|
Albrecht DS, Granziera C, Hooker JM, Loggia ML. In Vivo Imaging of Human Neuroinflammation. ACS Chem Neurosci 2016; 7:470-83. [PMID: 26985861 DOI: 10.1021/acschemneuro.6b00056] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is implicated in the pathophysiology of a growing number of human disorders, including multiple sclerosis, chronic pain, traumatic brain injury, and amyotrophic lateral sclerosis. As a result, interest in the development of novel methods to investigate neuroinflammatory processes, for the purpose of diagnosis, development of new therapies, and treatment monitoring, has surged over the past 15 years. Neuroimaging offers a wide array of non- or minimally invasive techniques to characterize neuroinflammatory processes. The intent of this Review is to provide brief descriptions of currently available neuroimaging methods to image neuroinflammation in the human central nervous system (CNS) in vivo. Specifically, because of the relatively widespread accessibility of equipment for nuclear imaging (positron emission tomography [PET]; single photon emission computed tomography [SPECT]) and magnetic resonance imaging (MRI), we will focus on strategies utilizing these technologies. We first provide a working definition of "neuroinflammation" and then discuss available neuroimaging methods to study human neuroinflammatory processes. Specifically, we will focus on neuroimaging methods that target (1) the activation of CNS immunocompetent cells (e.g. imaging of glial activation with TSPO tracer [(11)C]PBR28), (2) compromised BBB (e.g. identification of MS lesions with gadolinium-enhanced MRI), (3) CNS-infiltration of circulating immune cells (e.g. tracking monocyte infiltration into brain parenchyma with iron oxide nanoparticles and MRI), and (4) pathological consequences of neuroinflammation (e.g. imaging apoptosis with [(99m)Tc]Annexin V or iron accumulation with T2* relaxometry). This Review provides an overview of state-of-the-art techniques for imaging human neuroinflammation which have potential to impact patient care in the foreseeable future.
Collapse
Affiliation(s)
| | - Cristina Granziera
- Neuro-Immunology,
Neurology Division, Department of Clinical Neurosciences, Centre Hospitalier
Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
- LTS5, Ecole
Polytechnique
Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
38
|
Steinman MQ, Gao V, Alberini CM. The Role of Lactate-Mediated Metabolic Coupling between Astrocytes and Neurons in Long-Term Memory Formation. Front Integr Neurosci 2016; 10:10. [PMID: 26973477 PMCID: PMC4776217 DOI: 10.3389/fnint.2016.00010] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/15/2016] [Indexed: 01/07/2023] Open
Abstract
Long-term memory formation, the ability to retain information over time about an experience, is a complex function that affects multiple behaviors, and is an integral part of an individual's identity. In the last 50 years many scientists have focused their work on understanding the biological mechanisms underlying memory formation and processing. Molecular studies over the last three decades have mostly investigated, or given attention to, neuronal mechanisms. However, the brain is composed of different cell types that, by concerted actions, cooperate to mediate brain functions. Here, we consider some new insights that emerged from recent studies implicating astrocytic glycogen and glucose metabolisms, and particularly their coupling to neuronal functions via lactate, as an essential mechanism for long-term memory formation.
Collapse
Affiliation(s)
| | - Virginia Gao
- Center for Neural Science, New York University New York, NY, USA
| | | |
Collapse
|
39
|
Fryer RH, Bain J, De Vivo D. Mitochondrial Encephalomyopathy Lactic Acidosis and Stroke-Like Episodes (MELAS): A Case Report and Critical Reappraisal of Treatment Options. Pediatr Neurol 2016; 56:59-61. [PMID: 26797286 PMCID: PMC4789111 DOI: 10.1016/j.pediatrneurol.2015.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/15/2015] [Indexed: 01/31/2023]
Abstract
IMPORTANCE Stroke-like episodes signal progression and significant disability in the mitochondrial encephalomyopathy lactic acidosis and stroke-like episodes syndrome. Arginine is widely used as a treatment for stroke-like episode, although there is little evidence for this intervention. We discuss the management of a patient with mitochondrial encephalomyopathy lactic acidosis and stroke-like episodes who presented with a stroke-like episode. OBSERVATION During a seizure, which triggers the stroke-like episode, neurons are forced to utilize glycolysis as a source of adenosine triphosphate. Glycolytic by-products are damaging to the neuron. Breakdown of the blood-brain barrier leads to vasogenic edema. CONCLUSION Treatment of stroke-like episode should include anticonvulsants interictally to prevent seizures and dexamethasone ictally to help repair the blood-brain barrier.
Collapse
Affiliation(s)
- Robert H. Fryer
- Department of Neurology, Columbia University Medical Center, New York, NY,Corresponding Author: Robert H. Fryer, MD, PHD, 180 Fort Washington Avenue, Harkness 5th floor, New York, NY 10032, 212-342-2919 (P), 212-342-6865 (F),
| | - Jennifer Bain
- Department of Neurology, Columbia University Medical Center, New York, NY
| | - Darryl De Vivo
- Department of Neurology, Columbia University Medical Center, New York, NY
| |
Collapse
|
40
|
Grygorowicz T, Wełniak-Kamińska M, Strużyńska L. Early P2X7R-related astrogliosis in autoimmune encephalomyelitis. Mol Cell Neurosci 2016; 74:1-9. [PMID: 26921791 DOI: 10.1016/j.mcn.2016.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/22/2016] [Accepted: 02/22/2016] [Indexed: 01/16/2023] Open
Abstract
Astrocytes are the main cells responsible for maintenance of brain homeostasis. Undisturbed action and signaling with other cells are crucial for proper functioning of the central nervous system (CNS). Dysfunctional astrocytes may determine the degree of neuronal injury and are associated with several brain pathologies, among which are multiple sclerosis (MS) and the animal model of this disease which is known as experimental autoimmune encephalomyelitis (EAE). One of the many functions of astrocytes is their response to CNS damage when they undergo reactive gliosis. Our data reveal that activation of astrocytes occurs in forebrains of immunized rats at a very early stage of EAE, well before the symptomatic phase of the disease. We have noted enhanced expression of GFAP and S100β starting from day 4 post-immunization. Temporal coincidence between the expression of astrocyte activation markers and the expression of connexin 43 and purinergic P2X7 receptor (P2X7R) was also observed. Administration of Brilliant blue G, an antagonist of P2X7R, significantly decreases astrogliosis as confirmed by immunohistochemical analysis and observation of decreased levels of GFAP and S100β. The condition of the treated animals was improved and the neurological symptoms of the disease were alleviated. With the knowledge that cerebral astroglia represent the main source of ATP and glutamate which are potentially neurotoxic substances released through P2X7R and connexin hemichannels, we suggest that astroglia may be involved in pathogenesis of MS/EAE at a very early stage through the purinergic/glutamatergic mechanisms.
Collapse
Affiliation(s)
- Tomasz Grygorowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland.
| |
Collapse
|
41
|
Li CY, Wang YJ, Huang SW, Cheng CS, Wang HC. Replication of the Shrimp Virus WSSV Depends on Glutamate-Driven Anaplerosis. PLoS One 2016; 11:e0146902. [PMID: 26751681 PMCID: PMC4709008 DOI: 10.1371/journal.pone.0146902] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/23/2015] [Indexed: 12/14/2022] Open
Abstract
Infection with the white spot syndrome virus (WSSV) induces a metabolic shift in shrimp that resembles the "Warburg effect" in mammalian cells. This effect is triggered via activation of the PI3K-Akt-mTOR pathway, and it is usually accompanied by the activation of other metabolic pathways that provide energy and direct the flow of carbon and nitrogen. Here we show that unlike the glutamine metabolism (glutaminolysis) seen in most cancer cells to double deaminate glutamine to produce glutamate and the TCA cycle intermediate α-ketoglutarate (α-KG), at the WSSV genome replication stage (12 hpi), although glutaminase (GLS) expression was upregulated, only glutamate was taken up by the hemocytes of WSSV-infected shrimp. At the same time, we observed an increase in the activity of the two enzymes that convert glutamate to α-KG, glutamate dehydrogenase (GDH) and aspartate aminotransferase (ASAT). α-ketoglutarate concentration was also increased. A series of inhibition experiments suggested that the up-regulation of GDH is regulated by mTORC2, and that the PI3K-mTORC1 pathway is not involved. Suppression of GDH and ASAT by dsRNA silencing showed that both of these enzymes are important for WSSV replication. In GDH-silenced shrimp, direct replenishment of α-KG rescued both ATP production and WSSV replication. From these results, we propose a model of glutamate-driven anaplerosis that fuels the TCA cycle via α-KG and ultimately supports WSSV replication.
Collapse
Affiliation(s)
- Chun-Yuan Li
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Jan Wang
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Shiao-Wei Huang
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Cheng-Shun Cheng
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Han-Ching Wang
- Institute of Biotechnology, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
42
|
Monocarboxylate transporter 4 as a prognostic biomarker in patients with colorectal cancer and liver metastases. INTERNATIONAL JOURNAL OF SURGERY OPEN 2016. [DOI: 10.1016/j.ijso.2016.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Veyrat-Durebex C, Corcia P, Piver E, Devos D, Dangoumau A, Gouel F, Vourc'h P, Emond P, Laumonnier F, Nadal-Desbarats L, Gordon PH, Andres CR, Blasco H. Disruption of TCA Cycle and Glutamate Metabolism Identified by Metabolomics in an In Vitro Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2015; 53:6910-6924. [PMID: 26666663 DOI: 10.1007/s12035-015-9567-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/29/2015] [Indexed: 12/13/2022]
Abstract
This study aims to develop a cellular metabolomics model that reproduces the pathophysiological conditions found in amyotrophic lateral sclerosis in order to improve knowledge of disease physiology. We used a co-culture model combining the motor neuron-like cell line NSC-34 and the astrocyte clone C8-D1A, with each over-expressing wild-type or G93C mutant human SOD1, to examine amyotrophic lateral sclerosis (ALS) physiology. We focused on the effects of mutant human SOD1 as well as oxidative stress induced by menadione on intracellular metabolism using a metabolomics approach through gas chromatography coupled with mass spectrometry (GC-MS) analysis. Preliminary non-supervised analysis by Principal Component Analysis (PCA) revealed that cell type, genetic environment, and time of culture influenced the metabolomics profiles. Supervised analysis using orthogonal partial least squares discriminant analysis (OPLS-DA) on data from intracellular metabolomics profiles of SOD1G93C co-cultures produced metabolites involved in glutamate metabolism and the tricarboxylic acid cycle (TCA) cycle. This study revealed the feasibility of using a metabolomics approach in a cellular model of ALS. We identified potential disruption of the TCA cycle and glutamate metabolism under oxidative stress, which is consistent with prior research in the disease. Analysis of metabolic alterations in an in vitro model is a novel approach to investigation of disease physiology.
Collapse
Affiliation(s)
- Charlotte Veyrat-Durebex
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France. .,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France.
| | - Philippe Corcia
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Service de Neurologie, 37044, Tours, France
| | | | - David Devos
- Département de Pharmacologie médicale, INSERM U1171, Université Lille Nord de France, CHRU de Lille, Lille, France
| | - Audrey Dangoumau
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France
| | - Flore Gouel
- Département de Pharmacologie médicale, INSERM U1171, Université Lille Nord de France, CHRU de Lille, Lille, France
| | - Patrick Vourc'h
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France
| | - Patrick Emond
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,PPF-ASB, Université François Rabelais de Tours, Tours, France
| | - Frédéric Laumonnier
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France
| | - Lydie Nadal-Desbarats
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,PPF-ASB, Université François Rabelais de Tours, Tours, France
| | | | - Christian R Andres
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France
| | - Hélène Blasco
- UMR INSERM U930, Université François-Rabelais de Tours, Equipe « Neurogénétique et neurométabolomique », 37032, Tours, France.,CHRU de Tours, Laboratoire de Biochimie et de biologie moléculaire, 37044, Tours, France
| |
Collapse
|
44
|
Hong F, Sheng L, Ze Y, Hong J, Zhou Y, Wang L, Liu D, Yu X, Xu B, Zhao X, Ze X. Suppression of neurite outgrowth of primary cultured hippocampal neurons is involved in impairment of glutamate metabolism and NMDA receptor function caused by nanoparticulate TiO2. Biomaterials 2015; 53:76-85. [PMID: 25890708 DOI: 10.1016/j.biomaterials.2015.02.067] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/12/2015] [Accepted: 02/15/2015] [Indexed: 12/17/2022]
Abstract
Numerous studies have indicated that nano-titanium dioxide (TiO2) can induce neurotoxicity in vitro and in vivo, however, it is unclear whether nano-TiO2 affects neurite outgrowth of hippocampal neurons. In order to investigate the mechanism of neurotoxicity, rat primary cultured hippocampal neurons on the fourth day of culture were exposed to 5, 15, and 30 μg/mL nano-TiO2 for 24 h, and nano-TiO2 internalization, dendritic growth, glutamate metabolism, expression of N-methyl-D-aspartate (NMDA) receptor subunits (NR1, NR2A and NR2B), calcium homeostasis, sodium current (INa) and potassium current (IK) were examined. Our findings demonstrated that nano-TiO2 crossed the membrane into the cytoplasm or nucleus, and significantly suppressed dendritic growth of primary cultured hippocampal neurons in a concentration-dependent manner. Furthermore, nano-TiO2 induced a marked release of glutamate to the extracellular region, decreased glutamine synthetase activity and increased phosphate-activated glutaminase activity, elevated intracellular calcium ([Ca(2+)]i), down-regulated protein expression of NR1, NR2A and NR2B, and increased the amplitudes of the INa and IK. In addition, nano-TiO2 increased nitric oxide and nitrice synthase, attenuated the activities of Ca(2+)-ATPase and Na(+)/K(+)-ATPase, and increased the ADP/ATP ratio in the primary neurons. Taken together, these findings indicate that nano-TiO2 inhibits neurite outgrowth of hippocampal neurons by interfering with glutamate metabolism and impairing NMDA receptor function.
Collapse
Affiliation(s)
- Fashui Hong
- School of Life Science, Huaiyin Normal University, Huaian 223300, China.
| | - Lei Sheng
- Medical College of Soochow University, Suzhou 215123, China
| | - Yuguan Ze
- Medical College of Soochow University, Suzhou 215123, China
| | - Jie Hong
- Medical College of Soochow University, Suzhou 215123, China
| | - Yingjun Zhou
- School of Life Science, Huaiyin Normal University, Huaian 223300, China
| | - Ling Wang
- Library of Soochow University, Suzhou 215123, China
| | - Dong Liu
- Medical College of Soochow University, Suzhou 215123, China
| | - Xiaohong Yu
- Medical College of Soochow University, Suzhou 215123, China
| | - Bingqing Xu
- Medical College of Soochow University, Suzhou 215123, China
| | - Xiaoyang Zhao
- Medical College of Soochow University, Suzhou 215123, China
| | - Xiao Ze
- Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
45
|
Allaman I, Bélanger M, Magistretti PJ. Methylglyoxal, the dark side of glycolysis. Front Neurosci 2015; 9:23. [PMID: 25709564 PMCID: PMC4321437 DOI: 10.3389/fnins.2015.00023] [Citation(s) in RCA: 359] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/13/2015] [Indexed: 12/05/2022] Open
Abstract
Glucose is the main energy substrate for the brain. There is now extensive evidence indicating that the metabolic profile of neural cells with regard to glucose utilization and glycolysis rate is not homogenous, with a marked propensity for glycolytic glucose processing in astrocytes compared to neurons. Methylglyoxal, a highly reactive dicarbonyl compound, is inevitably formed as a by-product of glycolysis. Methylglyoxal is a major cell-permeant precursor of advanced glycation end-products (AGEs), which are associated with several pathologies including diabetes, aging and neurodegenerative diseases. In normal situations, cells are protected against methylglyoxal toxicity by different mechanisms and in particular the glyoxalase system, which represents the most important pathway for the detoxification of methylglyoxal. While the neurotoxic effects of methylglyoxal and AGEs are well characterized, our understanding the glyoxalase system in the brain is more scattered. Considering the high energy requirements (i.e., glucose) of the brain, one should expect that the cerebral glyoxalase system is adequately fitted to handle methylglyoxal toxicity. This review focuses on our actual knowledge on the cellular aspects of the glyoxalase system in brain cells, in particular with regard to its activity in astrocytes and neurons. A main emerging concept is that these two neural cell types have different and energetically adapted glyoxalase defense mechanisms which may serve as protective mechanism against methylglyoxal-induced cellular damage.
Collapse
Affiliation(s)
- Igor Allaman
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| | - Mireille Bélanger
- Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| | - Pierre J Magistretti
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology Thuwal, Saudi Arabia ; Laboratory of Neuroenergetics and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| |
Collapse
|
46
|
Bauernfeind AL, Babbitt CC. The appropriation of glucose through primate neurodevelopment. J Hum Evol 2014; 77:132-40. [DOI: 10.1016/j.jhevol.2014.05.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 03/22/2014] [Accepted: 05/02/2014] [Indexed: 12/25/2022]
|
47
|
Stromal-epithelial metabolic coupling in gastric cancer: Stromal MCT4 and mitochondrial TOMM20 as poor prognostic factors. Eur J Surg Oncol 2014; 40:1361-8. [DOI: 10.1016/j.ejso.2014.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/03/2014] [Accepted: 04/03/2014] [Indexed: 11/20/2022] Open
|
48
|
Abstract
Cerebral oedema (CO) after brain injury can occur from different ways. The vasogenic and cytotoxic oedema are usually described but osmotic and hydrostatic CO, respectively secondary to plasmatic hypotonia or increase in blood pressure, can also be encountered. Addition of these several mechanisms can worsen injuries. Consequences are major, leading quickly to death secondary to intracerebral hypertension and later to neuropsychic sequelae. So therapeutic care to control this phenomenon is essential and osmotherapy is actually the only way. A better understanding of physiopathological disorders, particularly energetic ways (lactate), aquaporine function, inflammation lead to new therapeutic hopes. The promising experimental results need now to be confirmed by clinical data.
Collapse
|
49
|
Lama S, Auer RN, Tyson R, Gallagher CN, Tomanek B, Sutherland GR. Lactate storm marks cerebral metabolism following brain trauma. J Biol Chem 2014; 289:20200-8. [PMID: 24849602 DOI: 10.1074/jbc.m114.570978] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain metabolism is thought to be maintained by neuronal-glial metabolic coupling. Glia take up glutamate from the synaptic cleft for conversion into glutamine, triggering glial glycolysis and lactate production. This lactate is shuttled into neurons and further metabolized. The origin and role of lactate in severe traumatic brain injury (TBI) remains controversial. Using a modified weight drop model of severe TBI and magnetic resonance (MR) spectroscopy with infusion of (13)C-labeled glucose, lactate, and acetate, the present study investigated the possibility that neuronal-glial metabolism is uncoupled following severe TBI. Histopathology of the model showed severe brain injury with subarachnoid and hemorrhage together with glial cell activation and positive staining for Tau at 90 min post-trauma. High resolution MR spectroscopy of brain metabolites revealed significant labeling of lactate at C-3 and C-2 irrespective of the infused substrates. Increased (13)C-labeled lactate in all study groups in the absence of ischemia implied activated astrocytic glycolysis and production of lactate with failure of neuronal uptake (i.e. a loss of glial sensing for glutamate). The early increase in extracellular lactate in severe TBI with the injured neurons rendered unable to pick it up probably contributes to a rapid progression toward irreversible injury and pan-necrosis. Hence, a method to detect and scavenge the excess extracellular lactate on site or early following severe TBI may be a potential primary therapeutic measure.
Collapse
Affiliation(s)
- Sanju Lama
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Roland N Auer
- the Hôpital Ste-Justine, Département de Pathologie, Université de Montréal, Montreal, Québec H3T 1C5, Canada
| | - Randy Tyson
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Clare N Gallagher
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Boguslaw Tomanek
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Garnette R Sutherland
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| |
Collapse
|
50
|
Ruiz M, Martínez-Vidal AF, Morales JM, Monleón D, Giménez Y Ribotta M. Neurodegenerative changes are prevented by Erythropoietin in the pmn model of motoneuron degeneration. Neuropharmacology 2014; 83:137-53. [PMID: 24769002 DOI: 10.1016/j.neuropharm.2014.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 03/02/2014] [Accepted: 04/10/2014] [Indexed: 11/29/2022]
Abstract
Motoneuron diseases are fatal neurodegenerative disorders characterized by a progressive loss of motoneurons, muscle weakness and premature death. The progressive motor neuronopathy (pmn) mutant mouse has been considered a good model for the autosomal recessive childhood form of spinal muscular atrophy (SMA). Here, we investigated the therapeutic potential of Erythropoietin (Epo) on this mutant mouse. Symptomatic or pre-symptomatic treatment with Epo significantly prolongs lifespan by 84.6% or 87.2% respectively. Epo preserves muscle strength and significantly attenuates behavioural motor deficits of mutant pmn mice. Histological and metabolic changes in the spinal cord evaluated by immunohistochemistry, western blot, and high-resolution (1)H-NMR spectroscopy were also greatly prevented by Epo-treatment. Our results illustrate the efficacy of Epo in improving quality of life of mutant pmn mice and open novel therapeutic pathways for motoneuron diseases.
Collapse
Affiliation(s)
- Marta Ruiz
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández (UMH), Av. Ramón y Cajal s/n, 03550 San Juan de Alicante, Alicante, Spain
| | - Ana Fe Martínez-Vidal
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández (UMH), Av. Ramón y Cajal s/n, 03550 San Juan de Alicante, Alicante, Spain
| | - José Manuel Morales
- Unidad Central de Investigación en Medicina, Universidad de Valencia, Valencia, Spain
| | - Daniel Monleón
- Fundación de Investigación del Hospital Clínico Universitario de Valencia (FIHCUV), Valencia, Spain
| | - Minerva Giménez Y Ribotta
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández (UMH), Av. Ramón y Cajal s/n, 03550 San Juan de Alicante, Alicante, Spain.
| |
Collapse
|