1
|
Zeng L, Yang J, Zhang C, Zhu J, Zhong S, Liu X, Xie H, Wang L, Chen L, Zhong M, Hua F, Liang W. Miro1: A potential target for treating neurological disorders. Neuroscience 2025; 577:228-239. [PMID: 40403957 DOI: 10.1016/j.neuroscience.2025.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 04/23/2025] [Accepted: 05/11/2025] [Indexed: 05/24/2025]
Abstract
The Miro1 protein is a member of the mitochondrial Rho GTPase (Miro) protein family and plays a crucial role in regulating the dynamic processes of mitochondria and participating in cellular movement and mitochondrial transport. In the nervous system, it ensures adequate energy supply for normal neuronal function and synaptic transmission. Additionally, Miro1 actively participates in the regulation of mitochondrial quality control and stress responses within neurons. Its primary function is to sense intracellular stress signals to regulate mitochondrial movement and metabolism, thereby adapting to environmental changes. Multiple studies have indicated that the Miro1 protein is associated with the pathogenesis of various neurological disorders, such as Alzheimer's Disease(AD), Parkinson's Disease(PD), and Amyotrophic Lateral Sclerosis(ALS). This article reviews the mechanistic role of Miro1 in these diseases and summarizes the latest research on its involvement in neurological disorders. These efforts aim to provide unified treatment strategies for certain neurological disorders and explore the potential for treating complex neurological diseases.
Collapse
Affiliation(s)
- Linghua Zeng
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China; Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Juan Yang
- Academic Affairs Department, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Conghui Zhang
- Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Junjie Zhu
- Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Saichun Zhong
- Department of Anesthesiology, Longnan First People's Hospital, Ganzhou 341000 Jiangxi, PR China.
| | - Xing Liu
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Haiyu Xie
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Lifeng Wang
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Li Chen
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Maolin Zhong
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang 330006 Jiangxi, PR China.
| | - Weidong Liang
- Anesthesia Surgery Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000 Jiangxi, PR China; Gannan Medical University, Ganzhou 341000 Jiangxi, PR China.
| |
Collapse
|
2
|
Fanibunda SE, Kukkemane K, Ghai U, Kolthur-Seetharam U, Hingorani L, Vaidya ADB, Vaidya VA. Withania somnifera Regulates Mitochondrial Biogenesis and Energetics in Rat Cortical Neurons: Role of BDNF and SIRT1. Mol Neurobiol 2025:10.1007/s12035-025-04920-7. [PMID: 40199807 DOI: 10.1007/s12035-025-04920-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Withania somnifera, a psychoactive plant with putative neuroprotective actions, is used in Indian traditional medicine for the treatment of neuropsychiatric and neurodegenerative disorders. However, the key mechanisms underlying the pleiotropic actions of Withania somnifera on the nervous system remain poorly understood. Given converging evidence suggests a critical role for mitochondrial dysfunction in the pathophysiology of neuropsychiatric and neurodegenerative diseases, we hypothesized that Withania somnifera may exert pleiotropic effects via targeting mitochondria. Treatment with Withania somnifera root extract (RE) or the withanolide-withanoside rich fraction (WLS) enhanced cellular ATP levels in rat cortical neurons in vitro and in the neocortex in vivo. In vivo respirometry performed on mitochondria isolated from the neocortex following RE or WLS treatment revealed increased mitochondrial respiration and OxPhos efficiency. Furthermore, WLS treatment evoked increases in mitochondrial mass, and RE and WLS treatments enhanced expression of brain derived neurotrophic factor (BDNF) and Sirtuin 1 (SIRT1), both in vitro and in vivo. Pharmacological inhibitor studies support an important role for BDNF and SIRT1 in the mitochondrial effects of Withania somnifera. Experiments with distinct phytochemical components of WLS identified withanolide A and withanoside IV as key constituents that enhance mitochondrial biogenesis and neuroenergetics. The neuroprotective actions of WLS, withanolide A and withanoside IV against corticosterone-induced neuronal cell death in vitro, required signaling via BDNF and SIRT1. Collectively, these results indicate that Withania somnifera root extract and specific phytochemical constituents robustly influence mitochondria in cortical neurons, contributing to stress adaptation and neuroprotection via BDNF and SIRT1 signaling.
Collapse
Affiliation(s)
- Sashaina E Fanibunda
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, 400005, India.
- Department of Reverse Pharmacology, Kasturba Health Society - Medical Research Centre, Khandubhai Desai Road, Vile Parle (W), Mumbai, 400056, India.
| | - Kowshik Kukkemane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, 400005, India
| | - Utkarsha Ghai
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, 400005, India
| | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, 400005, India
| | | | - Ashok D B Vaidya
- Department of Reverse Pharmacology, Kasturba Health Society - Medical Research Centre, Khandubhai Desai Road, Vile Parle (W), Mumbai, 400056, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, 400005, India.
| |
Collapse
|
3
|
K Soman S, Swain M, Dagda RK. BDNF-TrkB Signaling in Mitochondria: Implications for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:1756-1769. [PMID: 39030441 PMCID: PMC11909598 DOI: 10.1007/s12035-024-04357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a pivotal role in neuronal development, synaptic plasticity, and overall neuronal health by binding to its receptor, tyrosine receptor kinase B (TrkB). This review delves into the intricate mechanisms through which BDNF-TrkB signaling influences mitochondrial function and potentially influences pathology in neurodegenerative diseases. This review highlights the BDNF-TrkB signaling pathway which regulates mitochondrial bioenergetics, biogenesis, and dynamics, mitochondrial processes vital for synaptic transmission and plasticity. Furthermore, we explore how the BDNF-TrkB-PKA signaling in the cytosol and in mitochondria affects mitochondrial transport and distribution and mitochondrial content, which is crucial for supporting the energy demands of synapses. The dysregulation of this signaling pathway is linked to various neurodegenerative diseases, including Alzheimer's and Parkinson's disease, which are characterized by mitochondrial dysfunction and reduced BDNF expression. By examining seminal studies that have characterized this signaling pathway in health and disease, the present review underscores the potential of enhancing BDNF-TrkB signaling to mitigate mitochondrial dysfunction in neurodegenerative diseases, offering insights into therapeutic strategies to enhance neuronal resilience and function.
Collapse
Affiliation(s)
- Smijin K Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Ruben K Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA.
| |
Collapse
|
4
|
Zaninello M, Baptista P, Duarte FV. Mitochondrial Dynamics and mRNA Translation: A Local Synaptic Tale. BIOLOGY 2024; 13:746. [PMID: 39336173 PMCID: PMC11428642 DOI: 10.3390/biology13090746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Mitochondria are dynamic organelles that can adjust and respond to different stimuli within a cell. This plastic ability allows them to effectively coordinate several cellular functions in cells and becomes particularly relevant in highly complex cells such as neurons. An imbalance in mitochondrial dynamics can disrupt mitochondrial function, leading to abnormal cellular function and ultimately to a range of diseases, including neurodegenerative disorders. Regulation of mRNA transport and local translation inside neurons is crucial for maintaining the proteome of distal mitochondria, which is vital for energy production and synaptic function. A significant portion of the axonal transcriptome is dedicated to mRNAs for mitochondrial proteins, emphasizing the importance of local translation in sustaining mitochondrial function in areas far from the cell body. In neurons, local translation and the regulation of mRNAs encoding mitochondrial-shaping proteins could be essential for synaptic plasticity and neuronal health. The dynamics of these mRNAs, including their transport and local translation, may influence the morphology and function of mitochondria, thereby affecting the overall energy status and responsiveness of synapses. Comprehending the mitochondria-related mRNA regulation and local translation, as well as its influence on mitochondrial morphology near the synapses will help to better understand neuronal physiology and neurological diseases where mitochondrial dysfunction and impaired synaptic plasticity play a central role.
Collapse
Affiliation(s)
- Marta Zaninello
- Institute for Genetics, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Pedro Baptista
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filipe V Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
5
|
Sayehmiri F, Motamedi F, Batool Z, Naderi N, Shaerzadeh F, Zoghi A, Rezaei O, Khodagholi F, Pourbadie HG. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14897. [PMID: 39097920 PMCID: PMC11298206 DOI: 10.1111/cns.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
Synaptic plasticity is believed to underlie the cellular and molecular basis of memory formation. Mitochondria are one of the main organelles involved in metabolism and energy maintenance as plastic organelles that change morphologically and functionally in response to cellular needs and regulate synaptic function and plasticity through multiple mechanisms, including ATP generation, calcium homeostasis, and biogenesis. An increased neuronal activity enhances synaptic efficiency, during which mitochondria's spatial distribution and morphology change significantly. These organelles build up in the pre-and postsynaptic zones to produce ATP, which is necessary for several synaptic processes like neurotransmitter release and recycling. Mitochondria also regulate calcium homeostasis by buffering intracellular calcium, which ensures proper synaptic activity. Furthermore, mitochondria in the presynaptic terminal have distinct morphological properties compared to dendritic or postsynaptic mitochondria. This specialization enables precise control of synaptic activity and plasticity. Mitochondrial dysfunction has been linked to synaptic failure in many neurodegenerative disorders, like Alzheimer's disease (AD). In AD, malfunctioning mitochondria cause delays in synaptic vesicle release and recycling, ionic gradient imbalances, and mostly synaptic failure. This review emphasizes mitochondrial plasticity's contribution to synaptic function. It also explores the profound effect of mitochondrial malfunction on neurodegenerative disorders, focusing on AD, and provides an overview of how they sustain cellular health under normal conditions and how their malfunction contributes to neurodegenerative diseases, highlighting their potential as a therapeutic target for such conditions.
Collapse
Affiliation(s)
- Fatemeh Sayehmiri
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Nima Naderi
- Department of Pharmacology and Toxicology, Faculty of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | | | - Anahita Zoghi
- Department of Neurology, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Omidvar Rezaei
- Skull Base Research CenterLoghman Hakim Hospital, Shahid Beheshti University of Medical SciencesTehranIran
| | - Fariba Khodagholi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
6
|
Chen L, Zhang Y, Wang Z, Zhang Z, Wang J, Zhu G, Yang S. Activation of GPER1 by G1 prevents PTSD-like behaviors in mice: Illustrating the mechanisms from BDNF/TrkB to mitochondria and synaptic connection. CNS Neurosci Ther 2024; 30:e14855. [PMID: 38992889 PMCID: PMC11239537 DOI: 10.1111/cns.14855] [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: 04/12/2024] [Revised: 06/11/2024] [Accepted: 06/29/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND G1 is a specific agonist of G protein-coupled estrogen receptor 1 (GPER1), which binds and activates GPER1 to exert various neurological functions. However, the preventive effect of G1 on post-traumatic stress disorder (PTSD) and its mechanisms are unclear. OBJECTIVE To evaluate the protective effect of G1 against synaptic and mitochondrial impairments and to investigate the mechanism of G1 to improve PTSD from brain-derived neurotrophic factor (BDNF)/tyrosine kinase receptor B (TrkB) signaling. METHODS This study initially detected GPER1 expression in the hippocampus of single prolonged stress (SPS) mice, utilizing both Western blot and immunofluorescence staining. Subsequently, the effects of G1 on PTSD-like behaviors, synaptic, and mitochondrial functions in SPS mice were investigated. Additionally, the involvement of BDNF/TrkB signaling involved in the protection was further confirmed using GPER1 antagonist and TrkB inhibitor, respectively. RESULTS The expression of GPER1 was reduced in the hippocampus of SPS mice, and G1 treatment given for 14 consecutive days significantly improved PTSD-like behaviors in SPS mice compared with model group. Electrophysiological local field potential (LFP) results showed that G1 administration for 14 consecutive days could reverse the abnormal changes in the gamma oscillation in the CA1 region of SPS mice. Meanwhile, G1 administration for 14 consecutive days could significantly improve the abnormal expression of synaptic proteins, increase the expression of mitochondria-related proteins, increase the number of synapses in the hippocampus, and ameliorate the damage of hippocampal mitochondrial structure in SPS mice. In addition, G15 (GPER1 inhibitor) and ANA-12 (TrkB inhibitor) blocked the ameliorative effects of G1 on PTSD-like behaviors and aberrant expression of hippocampal synaptic and mitochondrial proteins in SPS mice and inhibited the reparative effects of G1 on structural damage to hippocampal mitochondria, respectively. CONCLUSION G1 improved PTSD-like behaviors in SPS mice, possibly by increasing hippocampal GPER1 expression and promoting BDNF/TrkB signaling to repair synaptic and mitochondrial functional impairments. This study would provide critical mechanism for the prevention and treatment of PTSD.
Collapse
Affiliation(s)
- Lixia Chen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Yang Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Zisheng Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Zhengrong Zhang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Shaojie Yang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
7
|
Gu Y, Pope A, Smith C, Carmona C, Johnstone A, Shi L, Chen X, Santos S, Bacon-Brenes CC, Shoff T, Kleczko KM, Frydman J, Thompson LM, Mobley WC, Wu C. BDNF and TRiC-inspired reagent rescue cortical synaptic deficits in a mouse model of Huntington's disease. Neurobiol Dis 2024; 195:106502. [PMID: 38608784 PMCID: PMC11890210 DOI: 10.1016/j.nbd.2024.106502] [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: 01/04/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Synaptic changes are early manifestations of neuronal dysfunction in Huntington's disease (HD). However, the mechanisms by which mutant HTT protein impacts synaptogenesis and function are not well understood. Herein we explored HD pathogenesis in the BACHD mouse model by examining synaptogenesis and function in long term primary cortical cultures. At DIV14 (days in vitro), BACHD cortical neurons showed no difference from WT neurons in synaptogenesis as revealed by colocalization of a pre-synaptic (Synapsin I) and a post-synaptic (PSD95) marker. From DIV21 to DIV35, BACHD neurons showed progressively reduced colocalization of Synapsin I and PSD95 relative to WT neurons. The deficits were effectively rescued by treatment of BACHD neurons with BDNF. The recombinant apical domain of CCT1 (ApiCCT1) yielded a partial rescuing effect. BACHD neurons also showed culture age-related significant functional deficits as revealed by multielectrode arrays (MEAs). These deficits were prevented by BDNF, whereas ApiCCT1 showed a less potent effect. These findings are evidence that deficits in BACHD synapse and function can be replicated in vitro and that BDNF or a TRiC-inspired reagent can potentially be protective against these changes in BACHD neurons. Our findings support the use of cellular models to further explicate HD pathogenesis and potential treatments.
Collapse
Affiliation(s)
- Yingli Gu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, 150001, China; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Alexander Pope
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Charlene Smith
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America
| | - Christopher Carmona
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America; Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, United States of America; Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Aaron Johnstone
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Linda Shi
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America; Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, United States of America; Beckman Laser Institute & Medical Clinic, University of California, Irvine, Irvine, CA, United States; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Xuqiao Chen
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Sarai Santos
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | | | - Thomas Shoff
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Korbin M Kleczko
- Department of Biology and Genetics, Stanford University, Stanford, CA 94305-5430, United States of America
| | - Judith Frydman
- Department of Biology and Genetics, Stanford University, Stanford, CA 94305-5430, United States of America
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, United States of America; Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, United States of America; Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States of America; Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697, United States of America
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, United States of America.
| |
Collapse
|
8
|
Pavlowsky A, Comyn T, Minatchy J, Geny D, Bun P, Danglot L, Preat T, Plaçais PY. Spaced training activates Miro/Milton-dependent mitochondrial dynamics in neuronal axons to sustain long-term memory. Curr Biol 2024; 34:1904-1917.e6. [PMID: 38642548 DOI: 10.1016/j.cub.2024.03.050] [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: 01/23/2023] [Revised: 12/21/2023] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
Neurons have differential and fluctuating energy needs across distinct cellular compartments, shaped by brain electrochemical activity associated with cognition. In vitro studies show that mitochondria transport from soma to axons is key to maintaining neuronal energy homeostasis. Nevertheless, whether the spatial distribution of neuronal mitochondria is dynamically adjusted in vivo in an experience-dependent manner remains unknown. In Drosophila, associative long-term memory (LTM) formation is initiated by an early and persistent upregulation of mitochondrial pyruvate flux in the axonal compartment of neurons in the mushroom body (MB). Through behavior experiments, super-resolution analysis of mitochondria morphology in the neuronal soma and in vivo mitochondrial fluorescence recovery after photobleaching (FRAP) measurements in the axons, we show that LTM induction, contrary to shorter-lived memories, is sustained by the departure of some mitochondria from MB neuronal soma and increased mitochondrial dynamics in the axonal compartment. Accordingly, impairing mitochondrial dynamics abolished the increased pyruvate consumption, specifically after spaced training and in the MB axonal compartment, thereby preventing LTM formation. Our results thus promote reorganization of the mitochondrial network in neurons as an integral step in elaborating high-order cognitive processes.
Collapse
Affiliation(s)
- Alice Pavlowsky
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Typhaine Comyn
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Julia Minatchy
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - David Geny
- Université de Paris, NeurImag Imaging Facility, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Philippe Bun
- Université de Paris, NeurImag Imaging Facility, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Lydia Danglot
- Université de Paris, NeurImag Imaging Facility, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Thomas Preat
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France.
| | - Pierre-Yves Plaçais
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France.
| |
Collapse
|
9
|
Wang CS, McCarthy CI, Guzikowski NJ, Kavalali ET, Monteggia LM. Brain-derived neurotrophic factor scales presynaptic calcium transients to modulate excitatory neurotransmission. Proc Natl Acad Sci U S A 2024; 121:e2303664121. [PMID: 38621124 PMCID: PMC11047077 DOI: 10.1073/pnas.2303664121] [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: 03/06/2023] [Accepted: 02/28/2024] [Indexed: 04/17/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays a critical role in synaptic physiology, as well as mechanisms underlying various neuropsychiatric diseases and their treatment. Despite its clear physiological role and disease relevance, BDNF's function at the presynaptic terminal, a fundamental unit of neurotransmission, remains poorly understood. In this study, we evaluated single synapse dynamics using optical imaging techniques in hippocampal cell cultures. We find that exogenous BDNF selectively increases evoked excitatory neurotransmission without affecting spontaneous neurotransmission. However, acutely blocking endogenous BDNF has no effect on evoked or spontaneous release, demonstrating that different approaches to studying BDNF may yield different results. When we suppressed BDNF-Tropomyosin receptor kinase B (TrkB) activity chronically over a period of days to weeks using a mouse line enabling conditional knockout of TrkB, we found that evoked glutamate release was significantly decreased while spontaneous release remained unchanged. Moreover, chronic blockade of BDNF-TrkB activity selectively downscales evoked calcium transients without affecting spontaneous calcium events. Via pharmacological blockade by voltage-gated calcium channel (VGCC) selective blockers, we found that the changes in evoked calcium transients are mediated by the P/Q subtype of VGCCs. These results suggest that BDNF-TrkB activity increases presynaptic VGCC activity to selectively increase evoked glutamate release.
Collapse
Affiliation(s)
- Camille S. Wang
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN3729-7933
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
| | - Clara I. McCarthy
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN3729-7933
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
| | - Natalie J. Guzikowski
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN3729-7933
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
| | - Ege T. Kavalali
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN3729-7933
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
| | - Lisa M. Monteggia
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN3729-7933
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240-7933
| |
Collapse
|
10
|
Lee HR, Choi SH, Lee SH. Differential involvement of mitochondria in post-tetanic potentiation at intracortical excitatory synapses of the medial prefrontal cortex. Cereb Cortex 2024; 34:bhad476. [PMID: 38061690 DOI: 10.1093/cercor/bhad476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/20/2022] [Accepted: 11/18/2023] [Indexed: 01/19/2024] Open
Abstract
Post-tetanic Ca2+ release from mitochondria produces presynaptic residual calcium, which contributes to post-tetanic potentiation. The loss of mitochondria-dependent post-tetanic potentiation is one of the earliest signs of Alzheimer's model mice. Post-tetanic potentiation at intracortical synapses of medial prefrontal cortex has been implicated in working memory. Although mitochondrial contribution to post-tetanic potentiation differs depending on synapse types, it is unknown which synapse types express mitochondria-dependent post-tetanic potentiation in the medial prefrontal cortex. We studied expression of mitochondria-dependent post-tetanic potentiation at different intracortical synapses of the rat medial prefrontal cortex. Post-tetanic potentiation occurred only at intracortical synapses onto layer 5 corticopontine cells from commissural cells and L2/3 pyramidal neurons. Among post-tetanic potentiation-expressing synapses, L2/3-corticopontine synapses in the prelimbic cortex were unique in that post-tetanic potentiation depends on mitochondria because post-tetanic potentiation at corresponding synapse types in other cortical areas was independent of mitochondria. Supporting mitochondria-dependent post-tetanic potentiation at L2/3-to-corticopontine synapses, mitochondria-dependent residual calcium at the axon terminals of L2/3 pyramidal neurons was significantly larger than that at commissural and corticopontine cells. Moreover, post-tetanic potentiation at L2/3-corticopontine synapses, but not at commissural-corticopontine synapses, was impaired in the young adult Alzheimer's model mice. These results would provide a knowledge base for comprehending synaptic mechanisms that underlies the initial clinical signs of neurodegenerative disorders.
Collapse
Affiliation(s)
- Hyoung-Ro Lee
- Department of Physiology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Hoon Choi
- Department of Physiology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Suk-Ho Lee
- Department of Physiology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
11
|
Toni M, Arena C, Cioni C, Tedeschi G. Temperature- and chemical-induced neurotoxicity in zebrafish. Front Physiol 2023; 14:1276941. [PMID: 37854466 PMCID: PMC10579595 DOI: 10.3389/fphys.2023.1276941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/22/2023] [Indexed: 10/20/2023] Open
Abstract
Throughout their lives, humans encounter a plethora of substances capable of inducing neurotoxic effects, including drugs, heavy metals and pesticides. Neurotoxicity manifests when exposure to these chemicals disrupts the normal functioning of the nervous system, and some neurotoxic agents have been linked to neurodegenerative pathologies such as Parkinson's and Alzheimer's disease. The growing concern surrounding the neurotoxic impacts of both naturally occurring and man-made toxic substances necessitates the identification of animal models for rapid testing across a wide spectrum of substances and concentrations, and the utilization of tools capable of detecting nervous system alterations spanning from the molecular level up to the behavioural one. Zebrafish (Danio rerio) is gaining prominence in the field of neuroscience due to its versatility. The possibility of analysing all developmental stages (embryo, larva and adult), applying the most common "omics" approaches (transcriptomics, proteomics, lipidomics, etc.) and conducting a wide range of behavioural tests makes zebrafish an excellent model for neurotoxicity studies. This review delves into the main experimental approaches adopted and the main markers analysed in neurotoxicity studies in zebrafish, showing that neurotoxic phenomena can be triggered not only by exposure to chemical substances but also by fluctuations in temperature. The findings presented here serve as a valuable resource for the study of neurotoxicity in zebrafish and define new scenarios in ecotoxicology suggesting that alterations in temperature can synergistically compound the neurotoxic effects of chemical substances, intensifying their detrimental impact on fish populations.
Collapse
Affiliation(s)
- Mattia Toni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Chiara Arena
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Carla Cioni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science (DIVAS), Università Degli Studi di Milano, Milano, Italy
- CRC “Innovation for Well-Being and Environment” (I-WE), Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
12
|
Aquilani R, Cotta Ramusino M, Maestri R, Iadarola P, Boselli M, Perini G, Boschi F, Dossena M, Bellini A, Buonocore D, Doria E, Costa A, Verri M. Several dementia subtypes and mild cognitive impairment share brain reduction of neurotransmitter precursor amino acids, impaired energy metabolism, and lipid hyperoxidation. Front Aging Neurosci 2023; 15:1237469. [PMID: 37655338 PMCID: PMC10466813 DOI: 10.3389/fnagi.2023.1237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Dementias and mild cognitive impairment (MCI) are associated with variously combined changes in the neurotransmitter system and signaling, from neurotransmitter synthesis to synaptic binding. The study tested the hypothesis that different dementia subtypes and MCI may share similar reductions of brain availability in amino acid precursors (AAPs) of neurotransmitter synthesis and concomitant similar impairment in energy production and increase of oxidative stress, i.e., two important metabolic alterations that impact neurotransmission. Materials and methods Sixty-five demented patients (Alzheimer's disease, AD, n = 44; frontotemporal disease, FTD, n = 13; vascular disease, VaD, n = 8), 10 subjects with MCI and 15 control subjects (CTRL) were recruited for this study. Cerebrospinal fluid (CSF) and plasma levels of AAPs, energy substrates (lactate, pyruvate), and an oxidative stress marker (malondialdehyde, MDA) were measured in all participants. Results Demented patients and subjects with MCI were similar for age, anthropometric parameters, biohumoral variables, insulin resistance (HOMA index model), and CSF neuropathology markers. Compared to age-matched CTRL, both demented patients and MCI subjects showed low CSF AAP tyrosine (precursor of dopamine and catecholamines), tryptophan (precursor of serotonin), methionine (precursor of acetylcholine) limited to AD and FTD, and phenylalanine (an essential amino acid largely used for protein synthesis) (p = 0.03 to <0.0001). No significant differences were found among dementia subtypes or between each dementia subtype and MCI subjects. In addition, demented patients and MCI subjects, compared to CTRL, had similar increases in CSF and plasma levels of pyruvate (CSF: p = 0.023 to <0.0001; plasma: p < 0.002 to <0.0001) and MDA (CSF: p < 0.035 to 0.002; plasma: p < 0.0001). Only in AD patients was the CSF level of lactate higher than in CTRL (p = 0.003). Lactate/pyruvate ratios were lower in all experimental groups than in CTRL. Conclusion AD, FTD, and VaD dementia patients and MCI subjects may share similar deficits in AAPs, partly in energy substrates, and similar increases in oxidative stress. These metabolic alterations may be due to AAP overconsumption following high brain protein turnover (leading to phenylalanine reductions), altered mitochondrial structure and function, and an excess of free radical production. All these metabolic alterations may have a negative impact on synaptic plasticity and activity.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Anna Bellini
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Enrico Doria
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
13
|
Duarte FV, Ciampi D, Duarte CB. Mitochondria as central hubs in synaptic modulation. Cell Mol Life Sci 2023; 80:173. [PMID: 37266732 DOI: 10.1007/s00018-023-04814-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/19/2023] [Indexed: 06/03/2023]
Abstract
Mitochondria are present in the pre- and post-synaptic regions, providing the energy required for the activity of these very specialized neuronal compartments. Biogenesis of synaptic mitochondria takes place in the cell body, and these organelles are then transported to the synapse by motor proteins that carry their cargo along microtubule tracks. The transport of mitochondria along neurites is a highly regulated process, being modulated by the pattern of neuronal activity and by extracellular cues that interact with surface receptors. These signals act by controlling the distribution of mitochondria and by regulating their activity. Therefore, mitochondria activity at the synapse allows the integration of different signals and the organelles are important players in the response to synaptic stimulation. Herein we review the available evidence regarding the regulation of mitochondrial dynamics by neuronal activity and by neuromodulators, and how these changes in the activity of mitochondria affect synaptic communication.
Collapse
Affiliation(s)
- Filipe V Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- III - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Daniele Ciampi
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
14
|
Savino R, Polito AN, Marsala G, Ventriglio A, Di Salvatore M, De Stefano MI, Valenzano A, Marinaccio L, Bellomo A, Cibelli G, Monda M, Monda V, Messina A, Polito R, Carotenuto M, Messina G. Agomelatine: A Potential Multitarget Compound for Neurodevelopmental Disorders. Brain Sci 2023; 13:734. [PMID: 37239206 PMCID: PMC10216109 DOI: 10.3390/brainsci13050734] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Agomelatine (AGM) is one of the latest atypical antidepressants, prescribed exclusively for the treatment of depression in adults. AGM belongs to the pharmaceutical class of melatonin agonist and selective serotonin antagonist ("MASS"), as it acts both as a selective agonist of melatonin receptors MT1 and MT2, and as a selective antagonist of 5-HT2C/5-HT2B receptors. AGM is involved in the resynchronization of interrupted circadian rhythms, with beneficial effects on sleep patterns, while antagonism on serotonin receptors increases the availability of norepinephrine and dopamine in the prefrontal cortex, with an antidepressant and nootropic effect. The use of AGM in the pediatric population is limited by the scarcity of data. In addition, few studies and case reports have been published on the use of AGM in patients with attention deficit and hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Considering this evidence, the purpose of this review is to report the potential role of AGM in neurological developmental disorders. AGM would increase the expression of the cytoskeleton-associated protein (ARC) in the prefrontal cortex, with optimization of learning, long-term memory consolidation, and improved survival of neurons. Another important feature of AGM is the ability to modulate glutamatergic neurotransmission in regions associated with mood and cognition. With its synergistic activity a melatoninergic agonist and an antagonist of 5-HT2C, AGM acts as an antidepressant, psychostimulant, and promoter of neuronal plasticity, regulating cognitive symptoms, resynchronizing circadian rhythms in patients with autism, ADHD, anxiety, and depression. Given its good tolerability and good compliance, it could potentially be administered to adolescents and children.
Collapse
Affiliation(s)
- Rosa Savino
- Department of Woman and Child, Neuropsychiatry for Child and Adolescent Unit, General Hospital "Riuniti" of Foggia, 71122 Foggia, Italy
| | - Anna Nunzia Polito
- Department of Woman and Child, Neuropsychiatry for Child and Adolescent Unit, General Hospital "Riuniti" of Foggia, 71122 Foggia, Italy
| | | | - Antonio Ventriglio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Melanie Di Salvatore
- Department of Woman and Child, Neuropsychiatry for Child and Adolescent Unit, General Hospital "Riuniti" of Foggia, 71122 Foggia, Italy
| | - Maria Ida De Stefano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Luigi Marinaccio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Antonello Bellomo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Vincenzo Monda
- Department of Movement Sciences and Wellbeing, University of Naples "Parthenope", 80133 Naples, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Rita Polito
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Marco Carotenuto
- Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
15
|
Liu QQ, Tian CJ, Li N, Chen ZC, Guo YL, Cheng DJ, Tang XY, Zhang XY. Brain-derived neurotrophic factor promotes airway smooth muscle cell proliferation in asthma through regulation of transient receptor potential channel-mediated autophagy. Mol Immunol 2023; 158:22-34. [PMID: 37094390 DOI: 10.1016/j.molimm.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE Increased proliferation of airway smooth muscle cells (ASMCs) is a key feature of airway remodeling in asthma. This study aims to determine whether brain-derived neurotrophic factor (BDNF) regulates ASMC proliferation and airway remodeling via the transient receptor potential channels (TRPCs)/autophagy axis. METHODS Human ASMCs were isolated and passively sensitized with human asthmatic serum. Protein levels of BDNF and its receptor TrkB, TRPC1/3/6, autophagy markers, intracellular Ca2+ concentration ([Ca2+]i), LC3 immunofluorescence, cell proliferation, cell cycle population were examined. Wistar rats were sensitized with OVA to establish asthma models. RESULTS In asthmatic serum-sensitized human ASMCs, BDNF overexpression or recombinant BDNF (rhBDNF) increased TrkB/TRPC1/3/6 axis, [Ca2+]i, autophagy level, cell proliferation, cell number in the S+G2/M phase and decreased cell number in the G0/G1 phase, whereas BDNF knockdown exerted the opposite effects. Furthermore, TRPC channel blocker SKF96365 and TRPC1/3/6 knockdown reversed the effects of the rhBDNF-mediated induction of [Ca2+]i, autophagy level, cell proliferation and cell number in the S+G2/M phase. Moreover, the autophagy inhibitor (3-MA) rescued the rhBDNF-mediated induction of cell proliferation and cell number in the S+G2/M phase. Further in vivo assays revealed that BDNF altered the pathology of airway remodeling, promoted the infiltration of inflammatory cells, promoted the proliferation of ASMCs, and upregulated the protein levels of TrkB, TRPC1/3/6, and autophagy markers in asthma model rats. CONCLUSION We conclude that BDNF promotes ASMCs proliferation in asthma through TRPC-mediated autophagy induction.
Collapse
Affiliation(s)
- Qian-Qian Liu
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Henan University, China
| | - Cui-Jie Tian
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China
| | - Nan Li
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China
| | - Zhuo-Chang Chen
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China
| | - Ya-Li Guo
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China
| | - Dong-Jun Cheng
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China
| | - Xue-Yi Tang
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China
| | - Xiao-Yu Zhang
- Department of Respiratory Disease and Intensive Care, Henan Provincial People's Hospital, China; Department of Respiratory Disease and Intensive Care, People's Hospital of Zhengzhou University, China.
| |
Collapse
|
16
|
Palasz E, Wilkaniec A, Stanaszek L, Andrzejewska A, Adamczyk A. Glia-Neurotrophic Factor Relationships: Possible Role in Pathobiology of Neuroinflammation-Related Brain Disorders. Int J Mol Sci 2023; 24:ijms24076321. [PMID: 37047292 PMCID: PMC10094105 DOI: 10.3390/ijms24076321] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Neurotrophic factors (NTFs) play an important role in maintaining homeostasis of the central nervous system (CNS) by regulating the survival, differentiation, maturation, and development of neurons and by participating in the regeneration of damaged tissues. Disturbances in the level and functioning of NTFs can lead to many diseases of the nervous system, including degenerative diseases, mental diseases, and neurodevelopmental disorders. Each CNS disease is characterized by a unique pathomechanism, however, the involvement of certain processes in its etiology is common, such as neuroinflammation, dysregulation of NTFs levels, or mitochondrial dysfunction. It has been shown that NTFs can control the activation of glial cells by directing them toward a neuroprotective and anti-inflammatory phenotype and activating signaling pathways responsible for neuronal survival. In this review, our goal is to outline the current state of knowledge about the processes affected by NTFs, the crosstalk between NTFs, mitochondria, and the nervous and immune systems, leading to the inhibition of neuroinflammation and oxidative stress, and thus the inhibition of the development and progression of CNS disorders.
Collapse
Affiliation(s)
- Ewelina Palasz
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: (E.P.); (A.A.)
| | - Anna Wilkaniec
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Luiza Stanaszek
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Anna Andrzejewska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Agata Adamczyk
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: (E.P.); (A.A.)
| |
Collapse
|
17
|
Fries GR, Saldana VA, Finnstein J, Rein T. Molecular pathways of major depressive disorder converge on the synapse. Mol Psychiatry 2023; 28:284-297. [PMID: 36203007 PMCID: PMC9540059 DOI: 10.1038/s41380-022-01806-1] [Citation(s) in RCA: 205] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 01/07/2023]
Abstract
Major depressive disorder (MDD) is a psychiatric disease of still poorly understood molecular etiology. Extensive studies at different molecular levels point to a high complexity of numerous interrelated pathways as the underpinnings of depression. Major systems under consideration include monoamines, stress, neurotrophins and neurogenesis, excitatory and inhibitory neurotransmission, mitochondrial dysfunction, (epi)genetics, inflammation, the opioid system, myelination, and the gut-brain axis, among others. This review aims at illustrating how these multiple signaling pathways and systems may interact to provide a more comprehensive view of MDD's neurobiology. In particular, considering the pattern of synaptic activity as the closest physical representation of mood, emotion, and conscience we can conceptualize, each pathway or molecular system will be scrutinized for links to synaptic neurotransmission. Models of the neurobiology of MDD will be discussed as well as future actions to improve the understanding of the disease and treatment options.
Collapse
Affiliation(s)
- Gabriel R. Fries
- grid.267308.80000 0000 9206 2401Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, TX 77054 USA ,grid.240145.60000 0001 2291 4776Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, TX 77030 USA
| | - Valeria A. Saldana
- grid.262285.90000 0000 8800 2297Frank H. Netter MD School of Medicine at Quinnipiac University, 370 Bassett Road, North Haven, CT 06473 USA
| | - Johannes Finnstein
- grid.419548.50000 0000 9497 5095Department of Translational Research in Psychiatry, Project Group Molecular Pathways of Depression, Max Planck Institute of Psychiatry, Kraepelinstr. 10, 80804 Munich, Germany
| | - Theo Rein
- Department of Translational Research in Psychiatry, Project Group Molecular Pathways of Depression, Max Planck Institute of Psychiatry, Kraepelinstr. 10, 80804, Munich, Germany.
| |
Collapse
|
18
|
Chen X, Shi L, Zhang L, Cheng Y, Xue Z, Yan J, Jiang H. Epitranscriptomic Analysis of N6-methyladenosine in Infant Rhesus Macaques after Multiple Sevoflurane Anesthesia. Neuroscience 2021; 482:64-76. [PMID: 34843896 DOI: 10.1016/j.neuroscience.2021.11.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 12/14/2022]
Abstract
Clinical investigations to date have proposed the possibility that exposure to anesthetics is associated with neurodevelopmental deficits. Sevoflurane is the most commonly used general anesthetic in pediatric patients. Animal studies have demonstrated that multiple exposures to sevoflurane during the postnatal period resulted in neuropathological brain changes and long-term cognitive deficits. However, the underlying mechanisms remain to be clarified. In this study, methylated RNA immunoprecipitation sequencing (MeRIP-Seq) was performed to acquire genome-wide profiling of RNA N6-methyladenosine (m6A) in the prefrontal cortex of infant rhesus macaques. The macaques in the sevoflurane group had more m6A peaks than the macaques in the control group (p ≤ 0.05). After sevoflurane treatment, the mRNA levels of YT521-B homology domain family 1 (YTHDF1) and YT521-B homology domain family 3 (YTHDF3) were decreased, and sevoflurane anesthesia dynamically regulated RNA m6A methylation. Gene ontology (GO) analysis revealed that after sevoflurane exposure, genes with increased methylation of m6A sites were enriched in some physiological processes relevant to neurodevelopment, mainly focused on synaptic plasticity. The female macaques had 18 hypermethylated genes. The males had 35 hypermethylated genes, and some physiological processes related to the regulation of synaptic structure were enriched. Rhesus macaques are genetically closer to human beings. Our findings can help in the study of the mechanism of sevoflurane-relevant neurodevelopmental deficits at the posttranscriptional level and can provide new insights into potential clinical preventions and interventions for the neurotoxicity of neonatal anesthesia exposure.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lingling Shi
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lei Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanyong Cheng
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhenyu Xue
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
19
|
Crombie GK, Palliser HK, Shaw JC, Hodgson DM, Walker DW, Hirst JJ. Neurosteroid-based intervention using Ganaxolone and Emapunil for improving stress-induced myelination deficits and neurobehavioural disorders. Psychoneuroendocrinology 2021; 133:105423. [PMID: 34601389 DOI: 10.1016/j.psyneuen.2021.105423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/11/2021] [Accepted: 09/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Prenatal stress is associated with long-term disturbances in white matter development and behaviour in children, such as attention deficit hyperactivity disorder (ADHD) and anxiety. Oligodendrocyte maturation and myelin formation is a tightly orchestrated process beginning during gestation, and therefore is very vulnerable to the effects of maternal prenatal stresses in mid-late pregnancy. The current study aimed to examine the effects of prenatal stress on components of the oligodendrocyte lineage to identify the key processes that are disrupted and to determine if postnatal therapies directed at ameliorating white matter deficits also improve behavioural outcomes. METHODS Pregnant guinea pig dams were exposed to control-handling or prenatal stress with strobe light exposure for 2hrs/day on gestational age (GA) 50, 55, 60 and 65, and allowed to spontaneously deliver ~GA70. Pups were administered oral ganaxolone (5 mg/kg/day in 45% cyclodextrin) or the TSPO agonist, emapunil (XBD173; 0.3 mg/kg/day in 1% tragacanth gum) or vehicle, on postnatal days (PND) 1-7. Behavioural outcomes were assessed using open field and elevated plus maze testing on PND7 and PND27. Hippocampal samples were collected at PND30 to assess markers of oligodendrocyte development through assessment of total oligodendrocytes (OLIG2) and mature cells (myelin basic protein; MBP), and total neurons (NeuN) by immunostaining. Real-time PCR was conducted on hippocampal regions to assess markers of the oligodendrocyte lineage, markers of neurogenesis and components of the neurosteroidogenesis pathway. Plasma samples were collected for steroid quantification of cortisol, allopregnanolone, progesterone and testosterone by ELISA. RESULTS Prenatal stress resulted in hyperactivity in male offspring, and anxiety-like behaviour in female offspring in the guinea pig at an age equivalent to late childhood. Postnatal ganaxolone and emapunil treatment after prenatal stress restored the behavioural phenotype to that of control in females only. The oligodendrocyte maturation lineage, translation of MBP mRNA-to-protein, and neurogenesis were disrupted in prenatally-stressed offspring, resulting in a decreased amount of mature myelin. Emapunil treatment restored mature myelin levels in both sexes, and reversed disruptions to the oligodendrocyte lineage in female offspring, an effect not seen with ganaxolone treatment. CONCLUSION The marked and persisting behavioural and white matter perturbations observed in a clinically relevant guinea pig model of prenatal stress highlights the need for postnatal interventions that increase myelin repair and improve long-term outcomes. The effectiveness of emapunil treatment in restoring female offspring behaviour, and promoting maturation of myelin indicates that early therapeutic interventions can reverse the damaging effects of major stressful events in pregnancy. Further studies optimising target mechanisms and dosing are warranted.
Collapse
Affiliation(s)
- Gabrielle K Crombie
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia.
| | - Hannah K Palliser
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| | - Julia C Shaw
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| | | | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, VIC, Australia
| | - Jonathan J Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| |
Collapse
|
20
|
Ahuja P, Ng CF, Pang BPS, Chan WS, Tse MCL, Bi X, Kwan HLR, Brobst D, Herlea-Pana O, Yang X, Du G, Saengnipanthkul S, Noh HL, Jiao B, Kim JK, Lee CW, Ye K, Chan CB. Muscle-generated BDNF (brain derived neurotrophic factor) maintains mitochondrial quality control in female mice. Autophagy 2021; 18:1367-1384. [PMID: 34689722 DOI: 10.1080/15548627.2021.1985257] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mitochondrial remodeling is dysregulated in metabolic diseases but the underlying mechanism is not fully understood. We report here that BDNF (brain derived neurotrophic factor) provokes mitochondrial fission and clearance in skeletal muscle via the PRKAA/AMPK-PINK1-PRKN/Parkin and PRKAA-DNM1L/DRP1-MFF pathways. Depleting Bdnf expression in myotubes reduced fatty acid-induced mitofission and mitophagy, which was associated with mitochondrial elongation and impaired lipid handling. Muscle-specific bdnf knockout (MBKO) mice displayed defective mitofission and mitophagy, and accumulation of dysfunctional mitochondria in the muscle when they were fed with a high-fat diet (HFD). These animals also have exacerbated body weight gain, increased intramyocellular lipid deposition, reduced energy expenditure, poor metabolic flexibility, and more insulin resistance. In contrast, consuming a BDNF mimetic (7,8-dihydroxyflavone) increased mitochondrial content, and enhanced mitofission and mitophagy in the skeletal muscles. Hence, BDNF is an essential myokine to maintain mitochondrial quality and function, and its repression in obesity might contribute to impaired metabolism.Abbreviation: 7,8-DHF: 7,8-dihydroxyflavone; ACACA/ACC: acetyl Coenzyme A carboxylase alpha; ACAD: acyl-Coenzyme A dehydrogenase family; ACADVL: acyl-Coenzyme A dehydrogenase, very long chain; ACOT: acyl-CoA thioesterase; CAMKK2: calcium/calmodulin-dependent protein kinase kinase 2, beta; BDNF: brain derived neurotrophic factor; BNIP3: BCL2/adenovirus E1B interacting protein 3; BNIP3L/NIX: BCL2/adenovirus E1B interacting protein 3-like; CCL2/MCP-1: chemokine (C-C motif) ligand 2; CCL5: chemokine (C-C motif) ligand 5; CNS: central nervous system; CPT1B: carnitine palmitoyltransferase 1b, muscle; Cpt2: carnitine palmitoyltransferase 2; CREB: cAMP responsive element binding protein; DNM1L/DRP1: dynamin 1-like; E2: estrogen; EHHADH: enoyl-CoenzymeA hydratase/3-hydroxyacyl CoenzymeA dehydrogenase; ESR1/ER-alpha: estrogen receptor 1 (alpha); FA: fatty acid; FAO: fatty acid oxidation; FCCP: carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone; FFA: free fatty acids; FGF21: fibroblast growth factor 21; FUNDC1: FUN14 domain containing 1; HADHA: hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha; HFD: high-fat diet; iWAT: inguinal white adipose tissues; MAP1LC3A/LC3A: microtubule-associated protein 1 light chain 3 alpha; MBKO; muscle-specific bdnf knockout; IL6/IL-6: interleukin 6; MCEE: methylmalonyl CoA epimerase; MFF: mitochondrial fission factor; NTRK2/TRKB: neurotrophic tyrosine kinase, receptor, type 2; OPTN: optineurin; PA: palmitic acid; PARL: presenilin associated, rhomboid-like; PDH: pyruvate dehydrogenase; PINK1: PTEN induced putative kinase 1; PPARGC1A/PGC-1α: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PRKAA/AMPK: protein kinase, AMP-activated, alpha 2 catalytic subunit; ROS: reactive oxygen species; TBK1: TANK-binding kinase 1; TG: triacylglycerides; TNF/TNFα: tumor necrosis factor; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1.
Collapse
Affiliation(s)
- Palak Ahuja
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Chun Fai Ng
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Brian Pak Shing Pang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Wing Suen Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Margaret Chui Ling Tse
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China, Hong Kong
| | - Xinyi Bi
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Hiu-Lam Rachel Kwan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China, Hong Kong
| | - Daniel Brobst
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Oana Herlea-Pana
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Suchaorn Saengnipanthkul
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Chi Wai Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China, Hong Kong
| | - Keqiang Ye
- Department of Pathology, Emory University School of Medicine, Atlanta, USA
| | - Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
| |
Collapse
|
21
|
Murali Mahadevan H, Hashemiaghdam A, Ashrafi G, Harbauer AB. Mitochondria in Neuronal Health: From Energy Metabolism to Parkinson's Disease. Adv Biol (Weinh) 2021; 5:e2100663. [PMID: 34382382 DOI: 10.1002/adbi.202100663] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/30/2021] [Indexed: 01/01/2023]
Abstract
Mitochondria are the main suppliers of neuronal adenosine triphosphate and play a critical role in brain energy metabolism. Mitochondria also serve as Ca2+ sinks and anabolic factories and are therefore essential for neuronal function and survival. Dysregulation of neuronal bioenergetics is increasingly implicated in neurodegenerative disorders, particularly Parkinson's disease. This review describes the role of mitochondria in energy metabolism under resting conditions and during synaptic transmission, and presents evidence for the contribution of neuronal mitochondrial dysfunction to Parkinson's disease.
Collapse
Affiliation(s)
| | - Arsalan Hashemiaghdam
- Department of Cell Biology and Physiology, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Angelika Bettina Harbauer
- Max-Planck-Institute for Neurobiology, 82152, Martinsried, Germany.,Technical University of Munich, Institute of Neuronal Cell Biology, 80333, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
22
|
Basu H, Pekkurnaz G, Falk J, Wei W, Chin M, Steen J, Schwarz TL. FHL2 anchors mitochondria to actin and adapts mitochondrial dynamics to glucose supply. J Cell Biol 2021; 220:212527. [PMID: 34342639 PMCID: PMC8340551 DOI: 10.1083/jcb.201912077] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/05/2021] [Accepted: 07/08/2021] [Indexed: 01/24/2023] Open
Abstract
Mitochondrial movement and distribution are fundamental to their function. Here we report a mechanism that regulates mitochondrial movement by anchoring mitochondria to the F-actin cytoskeleton. This mechanism is activated by an increase in glucose influx and the consequent O-GlcNAcylation of TRAK (Milton), a component of the mitochondrial motor-adaptor complex. The protein four and a half LIM domains protein 2 (FHL2) serves as the anchor. FHL2 associates with O-GlcNAcylated TRAK and is both necessary and sufficient to drive the accumulation of F-actin around mitochondria and to arrest mitochondrial movement by anchoring to F-actin. Disruption of F-actin restores mitochondrial movement that had been arrested by either TRAK O-GlcNAcylation or forced direction of FHL2 to mitochondria. This pathway for mitochondrial immobilization is present in both neurons and non-neuronal cells and can thereby adapt mitochondrial dynamics to changes in glucose availability.
Collapse
Affiliation(s)
- Himanish Basu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Division of Medical Sciences, Harvard Medical School, Boston, MA.,Department of Neurobiology, Harvard Medical School, Boston, MA
| | - Gulcin Pekkurnaz
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Department of Neurobiology, Harvard Medical School, Boston, MA
| | - Jill Falk
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Department of Neurobiology, Harvard Medical School, Boston, MA
| | - Wei Wei
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Morven Chin
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Division of Medical Sciences, Harvard Medical School, Boston, MA.,Department of Neurobiology, Harvard Medical School, Boston, MA
| | - Judith Steen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA
| | - Thomas L Schwarz
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA.,Department of Neurobiology, Harvard Medical School, Boston, MA
| |
Collapse
|
23
|
Lu D, Yu L, Li M, Zhai Q, Tian F, Chen W. Behavioral disorders caused by nonylphenol and strategies for protection. CHEMOSPHERE 2021; 275:129973. [PMID: 33639553 DOI: 10.1016/j.chemosphere.2021.129973] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/22/2021] [Accepted: 02/12/2021] [Indexed: 06/12/2023]
Abstract
Nonylphenol (NP) is widely used in daily production and life due to its good emulsification. In this review, we discuss toxicology studies that examined behavioral disorders caused by NP, the corresponding toxicological mechanisms in the central nervous system (CNS), and strategies for protection. Available in vitro and in vivo evidence suggests that exposure to NP during adulthood or early childhood is associated with cognitive dysfunction, including depression-like behaviors, anxiety-like behaviors, and impaired learning and memory. The main mechanisms underlying NP-related cognitive disorders include inflammation, destruction of synaptic plasticity, and destruction of important signaling pathways that affect the synthesis and secretion of neurotransmitters. The effects and mechanisms of NP exposure on CNS-mediated reproductive function, including interference with the expression of hormones, proteins, and enzymes, are discussed. Other abnormal behaviors such as locomotor activity and swimming behavior are also described. Several measures to prevent NP neurotoxicity are summarized. These measures are based on the toxicological mechanisms underlying NP exposure and include external protection and internal self-regulation of the nervous system. Finally, a new treatment idea is proposed based on the gut-brain axis. Characterizing the behavioral changes and underlying toxicity mechanisms associated with NP exposure and investigating the possible methods of treatment will help to expand the understanding of these mechanisms and could lead to more effective treatments.
Collapse
Affiliation(s)
- Dezhi Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, Jiangsu, 214122 China.
| | - Miaoyu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, Jiangsu, 214122 China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, Jiangsu, 214122 China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China; Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology & Business University, Beijing, 100048, China
| |
Collapse
|
24
|
The Axonal Glycolytic Pathway Contributes to Sensory Axon Extension and Growth Cone Dynamics. J Neurosci 2021; 41:6637-6651. [PMID: 34252036 DOI: 10.1523/jneurosci.0321-21.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/19/2021] [Accepted: 06/12/2021] [Indexed: 01/18/2023] Open
Abstract
Understanding the bioenergetics of axon extension and maintenance has wide ranging implications for neurodevelopment and disease states. Glycolysis is a pathway consisting of 10 enzymes and separated into preparatory and payoff phases, the latter producing ATP. Using embryonic chicken sensory neurons, we report that glycolytic enzymes are found through the axon and the growth cone. Pharmacological inhibition of glycolysis in the presence of NGF impairs axon extension and growth cone dynamics within minutes without affecting axon maintenance. Experiments using microfluidic chambers show that the effect of inhibiting glycolysis on axon extension is local along distal axons and can be reversed by promoting mitochondrial respiration. Knockdown of GAPDH simplifies growth cone morphology and is rescued by shRNA-resistant GAPDH expression. Rescue of GAPDH using KillerRed fused to GAPDH followed by localized chromophore-assisted light inactivation of KillerRed-GAPDH in distal axons halts growth cone dynamics. Considering filament polymerization requires ATP, inhibition of glycolysis results in a paradoxical increase in axonal actin filament levels. The effect on actin filaments is because of enzymes before GAPDH, the first enzyme in the payoff phase. In the absence of NGF, inhibition of glycolysis along distal axons results in axon degeneration independent of cell death. These data indicate that the glycolytic pathway is operative in distal axons and contributes to the rate of axon extension and growth cone dynamics in the presence of NGF and that, in the absence of NGF, the axonal glycolytic pathway is required for axon maintenance.SIGNIFICANCE STATEMENT Elucidation of the sources of ATP required for axon extension and maintenance has implications for understanding the mechanism of neuronal development and diseases of the nervous system. While recent work has emphasized the importance of mitochondrial oxidative phosphorylation, the role of the glycolytic pathway in axon morphogenesis and maintenance remains minimally understood. The data reveal that the glycolytic pathway is required for normal sensory axon extension in the presence of NGF, while in the absence of NGF the glycolytic pathway is required for axon maintenance. The results have implications for the understanding of the bioenergetics of axon morphogenesis and plasticity and indicate that NGF has protective effects on sensory axon maintenance in hypoglycemic states.
Collapse
|
25
|
Nishimura AL, Arias N. Synaptopathy Mechanisms in ALS Caused by C9orf72 Repeat Expansion. Front Cell Neurosci 2021; 15:660693. [PMID: 34140881 PMCID: PMC8203826 DOI: 10.3389/fncel.2021.660693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disease caused by degeneration of motor neurons (MNs). ALS pathogenic features include accumulation of misfolded proteins, glutamate excitotoxicity, mitochondrial dysfunction at distal axon terminals, and neuronal cytoskeleton changes. Synergies between loss of C9orf72 functions and gain of function by toxic effects of repeat expansions also contribute to C9orf72-mediated pathogenesis. However, the impact of haploinsufficiency of C9orf72 on neurons and in synaptic functions requires further examination. As the motor neurons degenerate, the disease symptoms will lead to neurotransmission deficiencies in the brain, spinal cord, and neuromuscular junction. Altered neuronal excitability, synaptic morphological changes, and C9orf72 protein and DPR localization at the synapses, suggest a potential involvement of C9orf72 at synapses. In this review article, we provide a conceptual framework for assessing the putative involvement of C9orf72 as a synaptopathy, and we explore the underlying and common disease mechanisms with other neurodegenerative diseases. Finally, we reflect on the major challenges of understanding C9orf72-ALS as a synaptopathy focusing on integrating mitochondrial and neuronal cytoskeleton degeneration as biomarkers and potential targets to treat ALS neurodegeneration.
Collapse
Affiliation(s)
- Agnes L Nishimura
- Department of Basic and Clinical Neuroscience, UK Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, UK Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
26
|
The Multifaceted Roles of Zinc in Neuronal Mitochondrial Dysfunction. Biomedicines 2021; 9:biomedicines9050489. [PMID: 33946782 PMCID: PMC8145363 DOI: 10.3390/biomedicines9050489] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
Zinc is a highly abundant cation in the brain, essential for cellular functions, including transcription, enzymatic activity, and cell signaling. However, zinc can also trigger injurious cascades in neurons, contributing to the pathology of neurodegenerative diseases. Mitochondria, critical for meeting the high energy demands of the central nervous system (CNS), are a principal target of the deleterious actions of zinc. An increasing body of work suggests that intracellular zinc can, under certain circumstances, contribute to neuronal damage by inhibiting mitochondrial energy processes, including dissipation of the mitochondrial membrane potential (MMP), leading to ATP depletion. Additional consequences of zinc-mediated mitochondrial damage include reactive oxygen species (ROS) generation, mitochondrial permeability transition, and excitotoxic calcium deregulation. Zinc can also induce mitochondrial fission, resulting in mitochondrial fragmentation, as well as inhibition of mitochondrial motility. Here, we review the known mechanisms responsible for the deleterious actions of zinc on the organelle, within the context of neuronal injury associated with neurodegenerative processes. Elucidating the critical contributions of zinc-induced mitochondrial defects to neurotoxicity and neurodegeneration may provide insight into novel therapeutic targets in the clinical setting.
Collapse
|
27
|
Chen Z, Xia Y, Liu H, Liu H, Xun L. The Mechanisms of Thiosulfate Toxicity against Saccharomyces cerevisiae. Antioxidants (Basel) 2021; 10:antiox10050646. [PMID: 33922196 PMCID: PMC8146336 DOI: 10.3390/antiox10050646] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 11/16/2022] Open
Abstract
Elemental sulfur and sulfite have been used to inhibit the growth of yeasts, but thiosulfate has not been reported to be toxic to yeasts. We observed that thiosulfate was more inhibitory than sulfite to Saccharomyces cerevisiae growing in a common yeast medium. At pH < 4, thiosulfate was a source of elemental sulfur and sulfurous acid, and both were highly toxic to the yeast. At pH 6, thiosulfate directly inhibited the electron transport chain in yeast mitochondria, leading to reductions in oxygen consumption, mitochondrial membrane potential and cellular ATP. Although thiosulfate was converted to sulfite and H2S by the mitochondrial rhodanese Rdl1, its toxicity was not due to H2S as the rdl1-deletion mutant that produced significantly less H2S was more sensitive to thiosulfate than the wild type. Evidence suggests that thiosulfate inhibits cytochrome c oxidase of the electron transport chain in yeast mitochondria. Thus, thiosulfate is a potential agent against yeasts.
Collapse
Affiliation(s)
- Zhigang Chen
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, China; (Z.C.); (Y.X.); (H.L.)
| | - Yongzhen Xia
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, China; (Z.C.); (Y.X.); (H.L.)
| | - Huaiwei Liu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, China; (Z.C.); (Y.X.); (H.L.)
| | - Honglei Liu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, China; (Z.C.); (Y.X.); (H.L.)
- Correspondence: (H.L.); (L.X.); Tel.: +86-15966642788 (H.L.); +1-509-335-2787 (L.X.)
| | - Luying Xun
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, China; (Z.C.); (Y.X.); (H.L.)
- School of Molecular Biosciences, Washington State University, Pullman, WA 991647520, USA
- Correspondence: (H.L.); (L.X.); Tel.: +86-15966642788 (H.L.); +1-509-335-2787 (L.X.)
| |
Collapse
|
28
|
Miller KM, Mercado NM, Sortwell CE. Synucleinopathy-associated pathogenesis in Parkinson's disease and the potential for brain-derived neurotrophic factor. NPJ PARKINSONS DISEASE 2021; 7:35. [PMID: 33846345 PMCID: PMC8041900 DOI: 10.1038/s41531-021-00179-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The lack of disease-modifying treatments for Parkinson’s disease (PD) is in part due to an incomplete understanding of the disease’s etiology. Alpha-synuclein (α-syn) has become a point of focus in PD due to its connection to both familial and idiopathic cases—specifically its localization to Lewy bodies (LBs), a pathological hallmark of PD. Within this review, we will present a comprehensive overview of the data linking synuclein-associated Lewy pathology with intracellular dysfunction. We first present the alterations in neuronal proteins and transcriptome associated with LBs in postmortem human PD tissue. We next compare these findings to those associated with LB-like inclusions initiated by in vitro exposure to α-syn preformed fibrils (PFFs) and highlight the profound and relatively unique reduction of brain-derived neurotrophic factor (BDNF) in this model. Finally, we discuss the multitude of ways in which BDNF offers the potential to exert disease-modifying effects on the basal ganglia. What remains unknown is the potential for BDNF to mitigate inclusion-associated dysfunction within the context of synucleinopathy. Collectively, this review reiterates the merit of using the PFF model as a tool to understand the physiological changes associated with LBs, while highlighting the neuroprotective potential of harnessing endogenous BDNF.
Collapse
Affiliation(s)
- Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA. .,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
29
|
Miao B, Yin Y, Mao G, Zhao B, Wu J, Shi H, Fei S. The implication of transient receptor potential canonical 6 in BDNF-induced mechanical allodynia in rat model of diabetic neuropathic pain. Life Sci 2021; 273:119308. [PMID: 33667520 DOI: 10.1016/j.lfs.2021.119308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 02/01/2023]
Abstract
AIMS Brain-derived neurotrophic factor (BDNF) is vital in the pathogenesis of mechanical allodynia with a paucity of reports available regarding diabetic neuropathy pain (DNP). Herein we identified the involvement of BDNF in driving mechanical allodynia in DNP rats via the activation of transient receptor potential canonical 6 (TRPC6) channel. MATERIALS AND METHODS The DNP rat model was established via streptozotocin (STZ) injection, and allodynia was assessed by paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL). The expression profiles of BDNF and TRPC6 in dorsal root ganglia (DRG) and spinal cord were illustrated by immunofluorescence and Western blotting. Intrathecal administration of K252a or TrkB-Fc was performed to inhibit BNDF/TrkB expression, and respective injection of GsMTX-4, BTP2 and TRPC6 antisense oligodeoxynucleotides (TRPC6-AS) was likewise conducted to inhibit TRPC6 expression in DNP rats. Calcium influx in DRG was monitored by calcium imaging. KEY FINDINGS The time-dependent increase of BDNF and TRPC6 expression in DRG and spinal cord was observed since the 7th post-STZ day, correlated with the development of mechanical allodynia in DNP rats. Intrathecal administration of K252a, TrkB-Fc, GsMTX-4 and BTP2 prevented mechanical allodynia in DNP rats. Pre-treatment of TRPC6-AS reversed the BDNF-induced pain-like responses in DNP rats rather than the naïve rats. In addition, the TRPC6-AS reversed BDNF-induced increase of calcium influx in DRG neurons in DNP rats. SIGNIFICANCE The intrathecal inhibition of TRPC6 alleviated the BDNF-induced mechanical allodynia in DNP rat model. This finding may validate the application of TRPC6 antagonists as interesting strategy for DNP management.
Collapse
Affiliation(s)
- Bei Miao
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China; Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Yue Yin
- Department of Anesthesiology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou 221009, Jiangsu Province, China
| | - Guangtong Mao
- Department of Pathology, Xinyi People's Hospital, 16 Renmin Road, Xinyi 221400, Jiangsu Province, China
| | - Benhuo Zhao
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Jiaojiao Wu
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Hengliang Shi
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China.
| | - Sujuan Fei
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China; Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China.
| |
Collapse
|
30
|
Neuroprotection by curcumin: A review on brain delivery strategies. Int J Pharm 2020; 585:119476. [DOI: 10.1016/j.ijpharm.2020.119476] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/05/2020] [Accepted: 05/24/2020] [Indexed: 12/26/2022]
|
31
|
Cheng H, Gang X, Liu Y, Wang G, Zhao X, Wang G. Mitochondrial dysfunction plays a key role in the development of neurodegenerative diseases in diabetes. Am J Physiol Endocrinol Metab 2020; 318:E750-E764. [PMID: 31714795 DOI: 10.1152/ajpendo.00179.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mitochondria have an essential function in cell survival due to their role in bioenergetics, reactive oxygen species generation, calcium buffering, and other metabolic activities. Mitochondrial dysfunctions are commonly found in neurodegenerative diseases (NDs), and diabetes is a risk factor for NDs. However, the role of mitochondria in diabetic neurodegeneration is still unclear. In the present study, we review the latest evidence on the role of mitochondrial dysfunctions in the development of diabetes-related NDs and the underlying molecular mechanisms. Hypoglycemic agents, especially metformin, have been proven to have neuroprotective effects in the treatment of diabetes, in which mitochondria could act as one of the underlying mechanisms. Other hypoglycemic agents, including thiazolidinediones (TZDs), dipeptidyl peptidase 4 (DPP-4) inhibitors, and glucagon-like peptide 1 (GLP-1) receptor agonists, have gained more attention because of their beneficial effects on NDs, presumably by improving mitochondrial function. Our review highlights the notion that mitochondria could be a promising therapeutic target in the treatment of NDs in patients with diabetes.
Collapse
Affiliation(s)
- Han Cheng
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Yujia Liu
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Gang Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
32
|
Chen G, Gan J, Yang C, Zuo Y, Peng J, Li M, Huo W, Xie Y, Zhang Y, Wang T, Deng X, Liang H. The SiaA/B/C/D signaling network regulates biofilm formation in Pseudomonas aeruginosa. EMBO J 2020; 39:e103412. [PMID: 32090355 DOI: 10.15252/embj.2019103412] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/21/2020] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
Bacterial cyclic-di-GMP (c-di-GMP) production is associated with biofilm development and the switch from acute to chronic infections. In Pseudomonas aeruginosa, the diguanylate cyclase (DGC) SiaD and phosphatase SiaA, which are co-transcribed as part of a siaABCD operon, are essential for cellular aggregation. However, the detailed functions of this operon and the relationships among its constituent genes are unknown. Here, we demonstrate that the siaABCD operon encodes for a signaling network that regulates SiaD enzymatic activity to control biofilm and aggregates formation. Through protein-protein interaction, SiaC promotes SiaD diguanylate cyclase activity. Biochemical and structural data revealed that SiaB is an unusual protein kinase that phosphorylates SiaC, whereas SiaA phosphatase can dephosphorylate SiaC. The phosphorylation state of SiaC is critical for its interaction with SiaD, which will switch on or off the DGC activity of SiaD and regulate c-di-GMP levels and subsequent virulence phenotypes. Collectively, our data provide insights into the molecular mechanisms underlying the modulation of DGC activity associated with chronic infections, which may facilitate the development of antimicrobial drugs.
Collapse
Affiliation(s)
- Gukui Chen
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jianhua Gan
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Chun Yang
- State Key Laboratory of Genetic Engineering, Shanghai Public Health Clinical Center, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yili Zuo
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Juan Peng
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Meng Li
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Weiping Huo
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yingpeng Xie
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yani Zhang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Tietao Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Haihua Liang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
33
|
Bagheri H, Ghasemi F, Barreto GE, Rafiee R, Sathyapalan T, Sahebkar A. Effects of curcumin on mitochondria in neurodegenerative diseases. Biofactors 2020; 46:5-20. [PMID: 31580521 DOI: 10.1002/biof.1566] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
Abstract
Neurodegenerative diseases (NDs) result from progressive deterioration of selectively susceptible neuron populations in different central nervous system (CNS) regions. NDs are classified in accordance with the primary clinical manifestations (e.g., parkinsonism, dementia, or motor neuron disease), the anatomic basis of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), and fundamental molecular abnormalities (e.g., mutations, mitochondrial dysfunction, and its related molecular alterations). NDs include the Alzheimer disease and Parkinson disease, among others. There is a growing evidence that mitochondrial dysfunction and its related mutations in the form of oxidative/nitrosative stress and neurotoxic compounds play major roles in the pathogenesis of various NDs. Curcumin, a polyphenol and nontoxic compound, obtained from turmeric, has been shown to have a therapeutic beneficial effect in various disorders especially on the CNS cells. It has been shown that curcumin has considerable neuro- and mitochondria-protective properties against broad-spectrum neurotoxic compounds and diseases/injury-associating NDs. In this article, we have reviewed the various effects of curcumin on mitochondrial dysfunction in NDs.
Collapse
Affiliation(s)
- Hossein Bagheri
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Rouhullah Rafiee
- Sciences and Research Branch, Islamic Azad University, Tehran, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
34
|
Jalilzad M, Jafari A, Babaei P. Neuregulin1β improves both spatial and associative learning and memory in Alzheimer model of rats possibly through signaling pathways other than Erk1/2. Neuropeptides 2019; 78:101963. [PMID: 31522857 DOI: 10.1016/j.npep.2019.101963] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Neuregulin-1β (NRG1 β) is associated with various neurological disorders such as schizophrenia, depression and Parkinson's disease. However, its role in Alzheimer's (AD) has not been understood yet. Here, we have studied the effect of NRG1 β and extracellular-signal-regulated kinase (ERK) signaling on special and associative memories and emotional stress in AD model of rats. METHODS Fifty six male Wistar rats were divided into eight groups of: Saline + Saline, Aβ + Saline, Aβ + NRG1β (5 μg/5 ul), Aβ + PBS, Aβ + NRG1β + PD98059 (PD, 5 μg/2 μl), Aβ + NRG1β + Saline and Saline + PD. AD model was induced by intracerebroventricular (ICV) injection of beta-amyloid protein (Aβ1-42, 4 μg/2 μl). The cognitive performances of rats were evaluated using Morris Water Maze (MWM) and Step through passive avoidance. Also locomotors activity and emotionality of animals were considered in an Open field test. Data were analyzed by one way Anova one way, repeated measure and T-test. RESULTS Significant improvement was found in spatial learning and memory assessed by total time spent in target quadrant [F (4, 32) = 12.4, p = 0.001], escape latency [F (4, 32) = 15.767, p = 0.001] and distance moved [F (4, 32) = 5.55, p = 0.002], in Aβ + NRG1β compared with Aβ + Saline in MWM. Also Aβ + NRG1β showed long latencies to enter into the dark compartment [F (4, 32) = 6.43, p = 0.001], but short time spent [F (4, 32) =6.93, p = 0.001] compared with control. Administration of an ERK inhibitor (PD98059, 5 μg, 15 min before NRG1β) didn't completely block learning memory restored by NRG1β in AD model (p = 0.7). No significant between groups differences was found in emotional stress characteristics in open field, except the grooming numbers which were higher in Saline + PD compared with Saline + Saline (p = 0.02). CONCLUSION Our findings indicate that NRG1β restores cognitive dysfunctions induced by amyloid β through signaling pathways possibly other than Erk1/2, with no significant change in anxiety, locomotion and vegetative activities.
Collapse
Affiliation(s)
- Marzieh Jalilzad
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvin Babaei
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
35
|
Khurram OU, Fogarty MJ, Sarrafian TL, Bhatt A, Mantilla CB, Sieck GC. Impact of aging on diaphragm muscle function in male and female Fischer 344 rats. Physiol Rep 2019; 6:e13786. [PMID: 29981218 PMCID: PMC6035336 DOI: 10.14814/phy2.13786] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/08/2018] [Accepted: 06/12/2018] [Indexed: 11/24/2022] Open
Abstract
The diaphragm muscle (DIAm) is the primary inspiratory muscle in mammals and is active during ventilatory behaviors, but it is also involved in higher-force behaviors such as those necessary for clearing the airway. Our laboratory has previously reported DIAm sarcopenia in rats and mice characterized by DIAm atrophy and a reduction in maximum specific force at 24 months of age. In Fischer 344 rats, these studies were limited to male animals, although in other studies, we noted a more rapid increase in body mass from 6 to 24 months of age in females (~140%) compared to males (~110%). This difference in body weight gain suggests a possible sex difference in the manifestation of sarcopenia. In mice, we previously measured transdiaphragmatic pressure (Pdi) to evaluate in vivo DIAm force generation across a range of motor behaviors, but found no evidence of sex-related differences. The purpose of this study in Fischer 344 rats was to evaluate if there are sex-related differences in DIAm sarcopenia, and if such differences translate to a functional impact on Pdi generation across motor behaviors and maximal Pdi (Pdimax ) elicited by bilateral phrenic nerve stimulation. In both males and females, DIAm sarcopenia was apparent in 24-month-old rats with a ~30% reduction in both maximum specific force and the cross-sectional area of type IIx and/or IIb fibers. Importantly, in both males and females, Pdi generated during ventilatory behaviors was unimpaired by sarcopenia, even during more forceful ventilatory efforts induced via airway occlusion. Although ventilatory behaviors were preserved with aging, there was a ~20% reduction in Pdimax , which likely impairs the ability of the DIAm to generate higher-force expulsive airway clearance behaviors necessary to maintain airway patency.
Collapse
Affiliation(s)
- Obaid U Khurram
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.,School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Tiffany L Sarrafian
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arjun Bhatt
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Carlos B Mantilla
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
36
|
Devine MJ, Kittler JT. Mitochondria at the neuronal presynapse in health and disease. Nat Rev Neurosci 2019; 19:63-80. [PMID: 29348666 DOI: 10.1038/nrn.2017.170] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Synapses enable neurons to communicate with each other and are therefore a prerequisite for normal brain function. Presynaptically, this communication requires energy and generates large fluctuations in calcium concentrations. Mitochondria are optimized for supplying energy and buffering calcium, and they are actively recruited to presynapses. However, not all presynapses contain mitochondria; thus, how might synapses with and without mitochondria differ? Mitochondria are also increasingly recognized to serve additional functions at the presynapse. Here, we discuss the importance of presynaptic mitochondria in maintaining neuronal homeostasis and how dysfunctional presynaptic mitochondria might contribute to the development of disease.
Collapse
Affiliation(s)
- Michael J Devine
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
37
|
Fields JA, Ellis RJ. HIV in the cART era and the mitochondrial: immune interface in the CNS. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 145:29-65. [PMID: 31208526 DOI: 10.1016/bs.irn.2019.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
HIV-associated neurocognitive disorders (HAND) persist in the era of effective combined antiretroviral therapy (cART). A large body of literature suggests that mitochondrial dysfunction is a prospective etiology of HAND in the cART era. While viral load is often suppressed and the immune system remains intact in HIV+ patients on cART, evidence suggests that the central nervous system (CNS) acts as a reservoir for virus and low-level expression of viral proteins, which interact with mitochondria. In particular, the HIV proteins glycoprotein 120, transactivator of transcription, viral protein R, and negative factor have each been linked to mitochondrial dysfunction in the brain. Moreover, cART drugs have also been shown to have detrimental effects on mitochondrial function. Here, we review the evidence generated from human studies, animal models, and in vitro models that support a role for HIV proteins and/or cART drugs in altered production of adenosine triphosphate, mitochondrial dynamics, mitophagy, calcium signaling and apoptosis, oxidative stress, mitochondrial biogenesis, and immunometabolism in the CNS. When insightful, evidence of HIV or cART-induced mitochondrial dysfunction in the peripheral nervous system or other cell types is discussed. Lastly, therapeutic approaches to targeting mitochondrial dysfunction have been summarized with the aim of guiding new investigations and providing hope that mitochondrial-based drugs may provide relief for those suffering with HAND.
Collapse
Affiliation(s)
- Jerel Adam Fields
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States.
| | - Ronald J Ellis
- Department of Neuroscience, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
38
|
Serotonin regulates mitochondrial biogenesis and function in rodent cortical neurons via the 5-HT 2A receptor and SIRT1-PGC-1α axis. Proc Natl Acad Sci U S A 2019; 116:11028-11037. [PMID: 31072928 PMCID: PMC6561197 DOI: 10.1073/pnas.1821332116] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neuronal mitochondria are crucial organelles that regulate bioenergetics and also modulate survival and function under environmental challenges. Here, we show that the neurotransmitter serotonin (5-HT) plays an important role in the making of new mitochondria (mitochondrial biogenesis) in cortical neurons, through the 5-HT2A receptor and via master regulators of mitochondrial biogenesis, SIRT1 and PGC-1α. Mitochondrial function is also enhanced by 5-HT, increasing cellular respiration and ATP, the energy currency of the cell. We found 5-HT reduces cellular reactive oxygen species and exerts potent neuroprotective action in neurons challenged with stress, an effect that requires SIRT1. These findings highlight a role for the mitochondrial effects of 5-HT in the facilitation of stress adaptation and identify drug targets to ameliorate mitochondrial dysfunction in neurons. Mitochondria in neurons, in addition to their primary role in bioenergetics, also contribute to specialized functions, including regulation of synaptic transmission, Ca2+ homeostasis, neuronal excitability, and stress adaptation. However, the factors that influence mitochondrial biogenesis and function in neurons remain poorly elucidated. Here, we identify an important role for serotonin (5-HT) as a regulator of mitochondrial biogenesis and function in rodent cortical neurons, via a 5-HT2A receptor-mediated recruitment of the SIRT1–PGC-1α axis, which is relevant to the neuroprotective action of 5-HT. We found that 5-HT increased mitochondrial biogenesis, reflected through enhanced mtDNA levels, mitotracker staining, and expression of mitochondrial components. This resulted in higher mitochondrial respiratory capacity, oxidative phosphorylation (OXPHOS) efficiency, and a consequential increase in cellular ATP levels. Mechanistically, the effects of 5-HT were mediated via the 5-HT2A receptor and master modulators of mitochondrial biogenesis, SIRT1 and PGC-1α. SIRT1 was required to mediate the effects of 5-HT on mitochondrial biogenesis and function in cortical neurons. In vivo studies revealed that 5-HT2A receptor stimulation increased cortical mtDNA and ATP levels in a SIRT1-dependent manner. Direct infusion of 5-HT into the neocortex and chemogenetic activation of 5-HT neurons also resulted in enhanced mitochondrial biogenesis and function in vivo. In cortical neurons, 5-HT enhanced expression of antioxidant enzymes, decreased cellular reactive oxygen species, and exhibited neuroprotection against excitotoxic and oxidative stress, an effect that required SIRT1. These findings identify 5-HT as an upstream regulator of mitochondrial biogenesis and function in cortical neurons and implicate the mitochondrial effects of 5-HT in its neuroprotective action.
Collapse
|
39
|
Li M, You M, Li S, Qiu Z, Wang Y. Effects of maternal exposure to nonylphenol on learning and memory in offspring involve inhibition of BDNF-PI3K/Akt signaling. Brain Res Bull 2019; 146:270-278. [PMID: 30660719 DOI: 10.1016/j.brainresbull.2019.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/21/2022]
Abstract
Nonylphenol (NP), a global environmental pollutant, has been found to result in impairments of neurodevelopment. However, effects of maternal exposure to NP on learning and memory and the potential mechanisms are largely unexplored. Thus, we treated dams with NP during gestation and lactation to study its effect on learning and memory in offspring. Morris water maze (MWM) task and the electrophysiological recording in the hippocampus were conducted in pups. We also investigated the activation of BDNF-PI3K/Akt signaling and the expression of its target protein PSD-95 in offspring hippocampus, which are curial for the synaptic plasticity and learning and memory. The results showed that maternal exposure to NP led to poor performance in MWM task and especially impairments of long-term potentiation (LTP), although the termination of NP exposure was at the end of lactation. Meanwhile, maternal exposure to NP also decreased the activation of BDNF-PI3K/Akt signaling and the protein level of PSD-95. Taken together, our results support the hypothesis that maternal exposure to NP during gestation and lactation causes damages to learning and memory. In addition, suppressed activation of the BDNF-PI3K/Akt signaling may contribute to these impairments caused by maternal exposure to NP.
Collapse
Affiliation(s)
- Mei Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Mingdan You
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Siyao Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Zhenmin Qiu
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, PR China.
| |
Collapse
|
40
|
Maynard KR, Hobbs JW, Phan BN, Gupta A, Rajpurohit S, Williams C, Rajpurohit A, Shin JH, Jaffe AE, Martinowich K. BDNF-TrkB signaling in oxytocin neurons contributes to maternal behavior. eLife 2018; 7:33676. [PMID: 30192229 PMCID: PMC6135608 DOI: 10.7554/elife.33676] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 09/02/2018] [Indexed: 12/18/2022] Open
Abstract
Brain-derived neurotrophic factor (Bdnf) transcription is controlled by several promoters, which drive expression of multiple transcripts encoding an identical protein. We previously reported that BDNF derived from promoters I and II is highly expressed in hypothalamus and is critical for regulating aggression in male mice. Here we report that BDNF loss from these promoters causes reduced sexual receptivity and impaired maternal care in female mice, which is concomitant with decreased oxytocin (Oxt) expression during development. We identify a novel link between BDNF signaling, oxytocin, and maternal behavior by demonstrating that ablation of TrkB selectively in OXT neurons partially recapitulates maternal care impairments observed in BDNF-deficient females. Using translating ribosome affinity purification and RNA-sequencing we define a molecular profile for OXT neurons and delineate how BDNF signaling impacts gene pathways critical for structural and functional plasticity. Our findings highlight BDNF as a modulator of sexually-dimorphic hypothalamic circuits that govern female-typical behaviors.
Collapse
Affiliation(s)
- Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - John W Hobbs
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - BaDoi N Phan
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - Amolika Gupta
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - Sumita Rajpurohit
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - Courtney Williams
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - Anandita Rajpurohit
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States.,Department of Mental Health, Johns Hopkins University, Baltimore, United States.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States.,Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, United States.,Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
41
|
Gershoni-Emek N, Altman T, Ionescu A, Costa CJ, Gradus-Pery T, Willis DE, Perlson E. Localization of RNAi Machinery to Axonal Branch Points and Growth Cones Is Facilitated by Mitochondria and Is Disrupted in ALS. Front Mol Neurosci 2018; 11:311. [PMID: 30233312 PMCID: PMC6134038 DOI: 10.3389/fnmol.2018.00311] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Local protein synthesis in neuronal axons plays an important role in essential spatiotemporal signaling processes; however, the molecular basis for the post-transcriptional regulation controlling this process in axons is still not fully understood. Here we studied the axonal mechanisms underlying the transport and localization of microRNA (miRNA) and the RNAi machinery along the axon. We first identified miRNAs, Dicer, and Argonaute-2 (Ago2) in motor neuron (MN) axons. We then studied the localization of RNAi machinery and demonstrated that mitochondria associate with miR-124 and RNAi proteins in axons. Importantly, this co-localization occurs primarily at axonal branch points and growth cones. Moreover, using live cell imaging of a functional Cy3-tagged miR-124, we revealed that this miRNA is actively transported with acidic compartments in axons, and associates with stalled mitochondria at growth cones and axonal branch points. Finally, we observed enhanced retrograde transport of miR-124-Cy3, and a reduction in its localization to static mitochondria in MNs expressing the ALS causative gene hSOD1G93A. Taken together, our data suggest that mitochondria participate in the axonal localization and transport of RNAi machinery, and further imply that alterations in this mechanism may be associated with neurodegeneration in ALS.
Collapse
Affiliation(s)
- Noga Gershoni-Emek
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Topaz Altman
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Ionescu
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Tal Gradus-Pery
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, United States.,Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Eran Perlson
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
42
|
Ferrari F, Gorini A, Hoyer S, Villa RF. Glutamate metabolism in cerebral mitochondria after ischemia and post-ischemic recovery during aging: relationships with brain energy metabolism. J Neurochem 2018; 146:416-428. [PMID: 29779216 DOI: 10.1111/jnc.14464] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022]
Abstract
Glutamate is involved in cerebral ischemic injury, but its role has not been completely clarified and studies are required to understand how to minimize its detrimental effects, contemporarily boosting the positive ones. In fact, glutamate is not only a neurotransmitter, but primarily a key metabolite for brain bioenergetics. Thus, we investigated the relationships between glutamate and brain energy metabolism in an in vivo model of complete cerebral ischemia of 15 min and during post-ischemic recovery after 1, 24, 48, 72, and 96 h in 1-year-old adult and 2-year-old aged rats. The maximum rates (Vmax ) of glutamate dehydrogenase (GlDH), glutamate-oxaloacetate transaminase, and glutamate-pyruvate transaminase were assayed in somatic mitochondria (FM) and in intra-synaptic 'Light' mitochondria and intra-synaptic 'Heavy' mitochondria ones purified from cerebral cortex, distinguishing post- and pre-synaptic compartments. During ischemia, none of the enzymes were modified in adult animals. In aged ones, glutamate-oxaloacetate transaminase was increased in FM and GlDH in intra-synaptic 'Heavy' mitochondria, stimulating glutamate catabolism. During post-ischemic recovery, FM did not show modifications at both ages while, in intra-synaptic mitochondria of adult animals, glutamate catabolism was increased after 1 h of recirculation and decreased after 48 and 72 h, whereas it remained decreased up to 96 h in aged rats. These results, with those previously published about Krebs' cycle and Electron Transport Chain (Villa et al., [2013] Neurochem. Int. 63, 765-781), demonstrate that: (i) Vmax of energy-linked enzymes are different in the various cerebral mitochondria, which (ii) respond differently to ischemia and post-ischemic recovery, also (iii) with respect to aging.
Collapse
Affiliation(s)
- Federica Ferrari
- Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Antonella Gorini
- Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Siegfried Hoyer
- Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.,Department of Pathology, University Clinic, University of Heidelberg, Heidelberg, Germany
| | - Roberto Federico Villa
- Laboratory of Pharmacology and Molecular Medicine of Central Nervous System, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
43
|
Akhtar MH, Hussain KK, Gurudatt NG, Chandra P, Shim YB. Ultrasensitive dual probe immunosensor for the monitoring of nicotine induced-brain derived neurotrophic factor released from cancer cells. Biosens Bioelectron 2018; 116:108-115. [PMID: 29860089 DOI: 10.1016/j.bios.2018.05.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/22/2018] [Accepted: 05/27/2018] [Indexed: 01/06/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) was detected in the extracellular matrix of neuronal cells using a dual probe immunosensor (DPI), where one of them was used as a working and another bioconjugate loading probe. The working probe was fabricated by covalently immobilizing capture anti-BDNF (Cap Ab) on the gold nanoparticles (AuNPs)/conducting polymer composite layer. The bioconjugate probe was modified by drop casting a bioconjugate particles composed of conducting polymer self-assembled AuNPs, immobilized with detection anti-BDNF (Det Ab) and toluidine blue O (TBO). Each sensor layer was characterized using the surface analysis and electrochemical methods. Two modified probes were precisely faced each other to form a microfluidic channel structure and the gap between inside modified surfaces was about 19 µm. At optimized conditions, the DPI showed a linear dynamic range from 4.0 to 600.0 pg/ml with a detection limit of 1.5 ± 0.012 pg/ml. Interference effect of IgG, arginine, glutamine, serine, albumin, and fibrinogene were examined and stability of the developed biosensor was also investigated. The reliability of the DPI sensor was evaluated by monitoring the extracellular release of BDNF using exogenic activators (ethanol, K+, and nicotine) in neuronal and non-neuronal cells. In addition, the effect of nicotine onto neuroblastoma cancer cells (SH-SY5Y) was studied in detail.
Collapse
Affiliation(s)
- Mahmood H Akhtar
- Department of Chemistry and Institute of BioPhysio Sensor Technology (IBST), Pusan National University, Busan 46241, South Korea
| | - Khalil K Hussain
- Department of Chemistry and Institute of BioPhysio Sensor Technology (IBST), Pusan National University, Busan 46241, South Korea
| | - N G Gurudatt
- Department of Chemistry and Institute of BioPhysio Sensor Technology (IBST), Pusan National University, Busan 46241, South Korea
| | - Pranjal Chandra
- Department of Chemistry and Institute of BioPhysio Sensor Technology (IBST), Pusan National University, Busan 46241, South Korea; Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Yoon-Bo Shim
- Department of Chemistry and Institute of BioPhysio Sensor Technology (IBST), Pusan National University, Busan 46241, South Korea.
| |
Collapse
|
44
|
Monday HR, Younts TJ, Castillo PE. Long-Term Plasticity of Neurotransmitter Release: Emerging Mechanisms and Contributions to Brain Function and Disease. Annu Rev Neurosci 2018; 41:299-322. [PMID: 29709205 DOI: 10.1146/annurev-neuro-080317-062155] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Long-lasting changes of brain function in response to experience rely on diverse forms of activity-dependent synaptic plasticity. Chief among them are long-term potentiation and long-term depression of neurotransmitter release, which are widely expressed by excitatory and inhibitory synapses throughout the central nervous system and can dynamically regulate information flow in neural circuits. This review article explores recent advances in presynaptic long-term plasticity mechanisms and contributions to circuit function. Growing evidence indicates that presynaptic plasticity may involve structural changes, presynaptic protein synthesis, and transsynaptic signaling. Presynaptic long-term plasticity can alter the short-term dynamics of neurotransmitter release, thereby contributing to circuit computations such as novelty detection, modifications of the excitatory/inhibitory balance, and sensory adaptation. In addition, presynaptic long-term plasticity underlies forms of learning and its dysregulation participates in several neuropsychiatric conditions, including schizophrenia, autism, intellectual disabilities, neurodegenerative diseases, and drug abuse.
Collapse
Affiliation(s)
- Hannah R Monday
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA;
| | - Thomas J Younts
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA;
| |
Collapse
|
45
|
|
46
|
Chen F, Danladi J, Ardalan M, Elfving B, Müller HK, Wegener G, Sanchez C, Nyengaard JR. A Critical Role of Mitochondria in BDNF-Associated Synaptic Plasticity After One-Week Vortioxetine Treatment. Int J Neuropsychopharmacol 2018; 21. [PMID: 29514282 PMCID: PMC6007239 DOI: 10.1093/ijnp/pyy022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Preclinical studies have indicated that antidepressant effect of vortioxetine involves increased synaptic plasticity and promotion of spine maturation. Mitochondria dysfunction may contribute to the pathophysiological basis of major depressive disorder. Taking into consideration that vortioxetine increases spine number and dendritic branching in hippocampus CA1 faster than fluoxetine, we hypothesize that new spines induced by vortioxetine can rapidly form functional synapses by mitochondrial support, accompanied by increased brain-derived neurotrophic factor signaling. METHODS Rats were treated for 1 week with vortioxetine or fluoxetine at pharmacologically relevant doses. Number of synapses and mitochondria in hippocampus CA1 were quantified by electron microscopy. Brain-derived neurotrophic factor protein levels were visualized with immunohistochemistry. Gene and protein expression of synapse and mitochondria-related markers were investigated with real-time quantitative polymerase chain reaction and immunoblotting. RESULTS Vortioxetine increased number of synapses and mitochondria significantly, whereas fluoxetine had no effect after 1-week dosing. BDNF levels in hippocampus DG and CA1 were significantly higher after vortioxetine treatment. Gene expression levels of Rac1 after vortioxetine treatment were significantly increased. There was a tendency towards increased gene expression levels of Drp1 and protein levels of Rac1. However, both gene and protein levels of c-Fos were significantly decreased. Furthermore, there was a significant positive correlation between BDNF levels and mitochondria and synapse numbers. CONCLUSION Our results imply that mitochondria play a critical role in synaptic plasticity accompanied by increased BDNF levels. Rapid changes in BDNF levels and synaptic/mitochondria plasticity of hippocampus following vortioxetine compared with fluoxetine may be ascribed to vortioxetine's modulation of serotonin receptors.
Collapse
Affiliation(s)
- Fenghua Chen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Correspondence: Fenghua Chen, Department of Clinical Medicine - Translational Neuropsychiatry Unit, Skovagervej 2, 8240 Risskov, Denmark ()
| | - Jibrin Danladi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Maryam Ardalan
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
- Department of Clinical Medicine - Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Heidi K Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
- Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
- AUGUST Centre, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
- Alkermes, Biotechnology, Waltham, MA
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Fogarty MJ, Omar TS, Zhan WZ, Mantilla CB, Sieck GC. Phrenic motor neuron loss in aged rats. J Neurophysiol 2018; 119:1852-1862. [PMID: 29412773 DOI: 10.1152/jn.00868.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is the age-related reduction of muscle mass and specific force. In previous studies, we found that sarcopenia of the diaphragm muscle (DIAm) is evident by 24 mo of age in both rats and mice and is associated with selective atrophy of type IIx and IIb muscle fibers and a decrease in maximum specific force. These fiber type-specific effects of sarcopenia resemble those induced by DIAm denervation, leading us to hypothesize that sarcopenia is due to an age-related loss of phrenic motor neurons (PhMNs). To address this hypothesis, we determined the number of PhMNs in young (6 mo old) and old (24 mo old) Fischer 344 rats. Moreover, we determined age-related changes in the size of PhMNs, since larger PhMNs innervate type IIx and IIb DIAm fibers. The PhMN pool was retrogradely labeled and imaged with confocal microscopy to assess the number of PhMNs and the morphometry of PhMN soma and proximal dendrites. In older animals, there were 22% fewer PhMNs, a 19% decrease in somal surface area, and a 21% decrease in dendritic surface area compared with young Fischer 344 rats. The age-associated loss of PhMNs involved predominantly larger PhMNs. These results are consistent with an age-related denervation of larger, more fatigable DIAm motor units, which are required primarily for high-force airway clearance behaviors. NEW & NOTEWORTHY Diaphragm muscle sarcopenia in rodent models is well described in the literature; however, the relationship between sarcopenia and frank phrenic motor neuron (MN) loss is unexplored in these models. We quantify a 22% loss of phrenic MNs in old (24 mo) compared with young (6 mo) Fischer 344 rats. We also report reductions in phrenic MN somal and proximal dendritic morphology that relate to decreased MN heterogeneity in old compared with young Fischer 344 rats.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,School of Biomedical Sciences, The University of Queensland , Brisbane , Australia
| | - Tanya S Omar
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Wen-Zhi Zhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Carlos B Mantilla
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Anesthesiology, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota.,Department of Anesthesiology, Mayo Clinic College of Medicine , Rochester, Minnesota
| |
Collapse
|
48
|
Liu W, Wang H, Xue X, Xia J, Liu J, Qi Z, Ji L. OGT-related mitochondrial motility is associated with sex differences and exercise effects in depression induced by prenatal exposure to glucocorticoids. J Affect Disord 2018; 226:203-215. [PMID: 28992584 DOI: 10.1016/j.jad.2017.09.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/19/2017] [Accepted: 09/27/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Prenatal exposure to glucocorticoids (GCs) has been found to trigger abnormal behaviors and deleterious neurological effects on offspring both in animals and in humans. The sex differences in depression have been replicated in numerous studies across cultures, persisting throughout the reproductive years. As an X-linked gene in rodents and in humans, O-GlcNAc transferase (OGT) may provide a novel perspective for the sex differences in depression. METHODS In the last third of pregnancy (gestational day 14-21), rats were subcutaneously administered either 0.13mg/kg dexamethasone-21-phosphate disodium salt (0.1mg/kg DEX) or vehicle (0.9% saline) once a day for 7 days. Adolescent (4 weeks) offspring were then trained in a swimming program or not. RESULTS Here we found that adult offspring rats exposed to DEX prenatally exhibited sex-specific depression-like behaviors, males being more vulnerable than females. Swimming exercise ameliorated the above-mentioned depressive syndromes, which may be a compensatory effect for male disadvantage suffering from prenatal stress. Furthermore, the effects of prenatal DEX exposure and swimming exercise on depression were associated with OGT-related mitochondrial motility, including PINK1/Parkin pathway and AKT/GSK3β pathway. LIMITATIONS Representative kymographs of mitochondrial motility were not detected and no causal effects were obtained by OGT gene overexpression or gene knockout in this study. CONCLUSIONS Our results provide a new perspective for better understanding sex differences and exercise effects in depression and may offer new mechanism-based therapeutic targets for depression.
Collapse
Affiliation(s)
- Weina Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Hongmei Wang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Xiangli Xue
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Jie Xia
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Jiatong Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Zhengtang Qi
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China.
| | - Liu Ji
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
49
|
Chen F, Ardalan M, Elfving B, Wegener G, Madsen TM, Nyengaard JR. Mitochondria Are Critical for BDNF-Mediated Synaptic and Vascular Plasticity of Hippocampus following Repeated Electroconvulsive Seizures. Int J Neuropsychopharmacol 2017; 21:291-304. [PMID: 29228215 PMCID: PMC5838811 DOI: 10.1093/ijnp/pyx115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 12/05/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Electroconvulsive therapy is a fast-acting and efficient treatment of depression used in the clinic. The underlying mechanism of its therapeutic effect is still unclear. However, recovery of synaptic connections and synaptic remodeling is thought to play a critical role for the clinical efficacy obtained from a rapid antidepressant response. Here, we investigated the relationship between synaptic changes and concomitant nonneuronal changes in microvasculature and mitochondria and its relationship to brain-derived neurotrophic factor level changes after repeated electroconvulsive seizures, an animal model of electroconvulsive therapy. METHODS Electroconvulsive seizures or sham treatment was given daily for 10 days to rats displaying a genetically driven phenotype modelling clinical depression: the Flinders Sensitive and Resistant Line rats. Stereological principles were employed to quantify numbers of synapses and mitochondria, and the length of microvessels in the hippocampus. The brain-derived neurotrophic factor protein levels were quantified with immunohistochemistry. RESULTS In untreated controls, a lower number of synapses and mitochondria was accompanied by shorter microvessels of the hippocampus in "depressive" phenotype (Flinders Sensitive Line) compared with the "nondepressed" phenotype (Flinders Resistant Line). Electroconvulsive seizure administration significantly increased the number of synapses and mitochondria, and length of microvessels both in Flinders Sensitive Line-electroconvulsive seizures and Flinders Resistant Line-electroconvulsive seizures rats. In addition, the amount of brain-derived neurotrophic factor protein was significantly increased in Flinders Sensitive Line and Flinders Resistant Line rats after electroconvulsive seizures. Furthermore, there was a significant positive correlation between brain-derived neurotrophic factor level and mitochondria/synapses. CONCLUSION Our results indicate that rapid and efficient therapeutic effect of electroconvulsive seizures may be related to synaptic plasticity, accompanied by brain-derived neurotrophic factor protein level elevation and mitochondrial and vascular support.
Collapse
Affiliation(s)
- Fenghua Chen
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Correspondence: Fenghua Chen, MD, PhD, Department of Clinical Medicine, Translational Neuropsychiatry Unit, Skovagervej 2, building 14K, 0.15, 8240 Risskov, Denmark ()
| | - Maryam Ardalan
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark,Center of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa,Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Gregers Wegener
- AUGUST Centre, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | | | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark,Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| |
Collapse
|
50
|
Smith GM, Gallo G. The role of mitochondria in axon development and regeneration. Dev Neurobiol 2017; 78:221-237. [PMID: 29030922 DOI: 10.1002/dneu.22546] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/12/2017] [Accepted: 10/08/2017] [Indexed: 12/26/2022]
Abstract
Mitochondria are dynamic organelles that undergo transport, fission, and fusion. The three main functions of mitochondria are to generate ATP, buffer cytosolic calcium, and generate reactive oxygen species. A large body of evidence indicates that mitochondria are either primary targets for neurological disease states and nervous system injury, or are major contributors to the ensuing pathologies. However, the roles of mitochondria in the development and regeneration of axons have just begun to be elucidated. Advances in the understanding of the functional roles of mitochondria in neurons had been largely impeded by insufficient knowledge regarding the molecular mechanisms that regulate mitochondrial transport, stalling, fission/fusion, and a paucity of approaches to image and analyze mitochondria in living axons at the level of the single mitochondrion. However, technical advances in the imaging and analysis of mitochondria in living neurons and significant insights into the mechanisms that regulate mitochondrial dynamics have allowed the field to advance. Mitochondria have now been attributed important roles in the mechanism of axon extension, regeneration, and axon branching. The availability of new experimental tools is expected to rapidly increase our understanding of the functions of axonal mitochondria during both development and later regenerative attempts. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 221-237, 2018.
Collapse
Affiliation(s)
- George M Smith
- Department of Neuroscience, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140.,Shriners Hospitals Pediatric Research Center, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Gianluca Gallo
- Department of Anatomy and Cell Biology, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140.,Shriners Hospitals Pediatric Research Center, Temple University, Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| |
Collapse
|