51
|
Sabharwal V, Koushika SP. Crowd Control: Effects of Physical Crowding on Cargo Movement in Healthy and Diseased Neurons. Front Cell Neurosci 2019; 13:470. [PMID: 31708745 PMCID: PMC6823667 DOI: 10.3389/fncel.2019.00470] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/02/2019] [Indexed: 01/22/2023] Open
Abstract
High concentration of cytoskeletal filaments, organelles, and proteins along with the space constraints due to the axon's narrow geometry lead inevitably to intracellular physical crowding along the axon of a neuron. Local cargo movement is essential for maintaining steady cargo transport in the axon, and this may be impeded by physical crowding. Molecular motors that mediate active transport share movement mechanisms that allow them to bypass physical crowding present on microtubule tracks. Many neurodegenerative diseases, irrespective of how they are initiated, show increased physical crowding owing to the greater number of stalled organelles and structural changes associated with the cytoskeleton. Increased physical crowding may be a significant factor in slowing cargo transport to synapses, contributing to disease progression and culminating in the dying back of the neuronal process. This review explores the idea that physical crowding can impede cargo movement along the neuronal process. We examine the sources of physical crowding and strategies used by molecular motors that might enable cargo to circumvent physically crowded locations. Finally, we describe sub-cellular changes in neurodegenerative diseases that may alter physical crowding and discuss the implications of such changes on cargo movement.
Collapse
Affiliation(s)
| | - Sandhya P. Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
52
|
Bordone MP, Salman MM, Titus HE, Amini E, Andersen JV, Chakraborti B, Diuba AV, Dubouskaya TG, Ehrke E, Espindola de Freitas A, Braga de Freitas G, Gonçalves RA, Gupta D, Gupta R, Ha SR, Hemming IA, Jaggar M, Jakobsen E, Kumari P, Lakkappa N, Marsh APL, Mitlöhner J, Ogawa Y, Paidi RK, Ribeiro FC, Salamian A, Saleem S, Sharma S, Silva JM, Singh S, Sulakhiya K, Tefera TW, Vafadari B, Yadav A, Yamazaki R, Seidenbecher CI. The energetic brain - A review from students to students. J Neurochem 2019; 151:139-165. [PMID: 31318452 DOI: 10.1111/jnc.14829] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022]
Abstract
The past 20 years have resulted in unprecedented progress in understanding brain energy metabolism and its role in health and disease. In this review, which was initiated at the 14th International Society for Neurochemistry Advanced School, we address the basic concepts of brain energy metabolism and approach the question of why the brain has high energy expenditure. Our review illustrates that the vertebrate brain has a high need for energy because of the high number of neurons and the need to maintain a delicate interplay between energy metabolism, neurotransmission, and plasticity. Disturbances to the energetic balance, to mitochondria quality control or to glia-neuron metabolic interaction may lead to brain circuit malfunction or even severe disorders of the CNS. We cover neuronal energy consumption in neural transmission and basic ('housekeeping') cellular processes. Additionally, we describe the most common (glucose) and alternative sources of energy namely glutamate, lactate, ketone bodies, and medium chain fatty acids. We discuss the multifaceted role of non-neuronal cells in the transport of energy substrates from circulation (pericytes and astrocytes) and in the supply (astrocytes and microglia) and usage of different energy fuels. Finally, we address pathological consequences of disrupted energy homeostasis in the CNS.
Collapse
Affiliation(s)
- Melina Paula Bordone
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mootaz M Salman
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Haley E Titus
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elham Amini
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Artem V Diuba
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatsiana G Dubouskaya
- Institute of Biophysics and Cell Engineering, National Academy of Sciences of Belarus, Minsk, Belarus
| | - Eric Ehrke
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Andiara Espindola de Freitas
- Neurobiology Section, Biological Sciences Division, University of California, San Diego, La Jolla, California, USA
| | | | | | | | - Richa Gupta
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Sharon R Ha
- Baylor College of Medicine, Houston, Texas, USA
| | - Isabel A Hemming
- Brain Growth and Disease Laboratory, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,School of Medicine and Pharmacology, The University of Western Australia, Crawley, Australia
| | - Minal Jaggar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Punita Kumari
- Defense Institute of Physiology and allied sciences, Defense Research and Development Organization, Timarpur, Delhi, India
| | - Navya Lakkappa
- Department of Pharmacology, JSS college of Pharmacy, Ooty, India
| | - Ashley P L Marsh
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jessica Mitlöhner
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Yuki Ogawa
- The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | | | | | - Ahmad Salamian
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Suraiya Saleem
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sorabh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Joana M Silva
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal
| | - Shripriya Singh
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Kunjbihari Sulakhiya
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Tesfaye Wolde Tefera
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Behnam Vafadari
- Institute of environmental medicine, UNIKA-T, Technical University of Munich, Munich, Germany
| | - Anuradha Yadav
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Reiji Yamazaki
- Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.,Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University, Magdeburg, Germany
| | - Constanze I Seidenbecher
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
53
|
Fractionated mitochondrial magnetic separation for isolation of synaptic mitochondria from brain tissue. Sci Rep 2019; 9:9656. [PMID: 31273236 PMCID: PMC6609636 DOI: 10.1038/s41598-019-45568-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022] Open
Abstract
While mitochondria maintain essential cellular functions, such as energy production, calcium homeostasis, and regulating programmed cellular death, they also play a major role in pathophysiology of many neurological disorders. Furthermore, several neurodegenerative diseases are closely linked with synaptic damage and synaptic mitochondrial dysfunction. Unfortunately, the ability to assess mitochondrial dysfunction and the efficacy of mitochondrial-targeted therapies in experimental models of neurodegenerative disease and CNS injury is limited by current mitochondrial isolation techniques. Density gradient ultracentrifugation (UC) is currently the only technique that can separate synaptic and non-synaptic mitochondrial sub-populations, though small brain regions cannot be assayed due to low mitochondrial yield. To address this limitation, we used fractionated mitochondrial magnetic separation (FMMS), employing magnetic anti-Tom22 antibodies, to develop a novel strategy for isolation of functional synaptic and non-synaptic mitochondria from mouse cortex and hippocampus without the usage of UC. We compared the yield and functionality of mitochondria derived using FMMS to those derived by UC. FMMS produced 3x more synaptic mitochondrial protein yield compared to UC from the same amount of tissue, a mouse hippocampus. FMMS also has increased sensitivity, compared to UC separation, to measure decreased mitochondrial respiration, demonstrated in a paradigm of mild closed head injury. Taken together, FMMS enables improved brain-derived mitochondrial yield for mitochondrial assessments and better detection of mitochondrial impairment in CNS injury and neurodegenerative disease.
Collapse
|
54
|
TSPO upregulation in bipolar disorder and concomitant downregulation of mitophagic proteins and NLRP3 inflammasome activation. Neuropsychopharmacology 2019; 44:1291-1299. [PMID: 30575805 PMCID: PMC6785146 DOI: 10.1038/s41386-018-0293-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/24/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Abstract
Bipolar disorder (BD) is a chronic, debilitating illness with a global prevalence of up to 4.8%. The importance of understanding how dysfunctional mitochondria and mitophagy contribute to cell survival and death in BD is becoming increasingly apparent. Therefore, the purpose of this study was to evaluate the mitophagic pathway and NLRP3 inflammasome activation in peripheral blood mononuclear cells (PBMCs) of patients with BD and healthy individuals. Since 18-kDa translocator protein (TSPO) plays an important role in regulating mitochondrial function and since TSPO itself impairs cellular mitophagy, we also investigated the changes in the TSPO-related pathway. Our results showed that patients with BD had lower levels of Parkin, p62/SQSTM1 and LC3A and an upregulation of TSPO pathway proteins (TSPO and VDAC), both in terms of mRNA and protein levels. Additionally, we found a negative correlation between mitophagy-related proteins and TSPO levels, while VDAC correlated negatively with p62/SQSTM1 and LC3 protein levels. Moreover, we found that the gene expression levels of the NLRP3-related proteins NLRP3, ASC, and pro-casp1 were upregulated in BD patients, followed by an increase in caspase-1 activity as well as IL-1β and IL-18 levels. As expected, there was a strong positive correlation between NLRP3-related inflammasome activation and TSPO-related proteins. The data reported here suggest that TSPO-VDAC complex upregulation in BD patients, the simultaneous downregulation of mitophagic proteins and NLRP3 inflammasome activation could lead to an accumulation of dysfunctional mitochondria, resulting in inflammation and apoptosis. In summary, the findings of this study provide novel evidence that mitochondrial dysfunction measured in peripheral blood is associated with BD.
Collapse
|
55
|
Hayes LR, Asress SA, Li Y, Galkin A, Stepanova A, Kawamata H, Manfredi G, Glass JD. Distal denervation in the SOD1 knockout mouse correlates with loss of mitochondria at the motor nerve terminal. Exp Neurol 2019; 318:251-257. [PMID: 31082391 DOI: 10.1016/j.expneurol.2019.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/03/2019] [Accepted: 05/08/2019] [Indexed: 12/01/2022]
Abstract
Impairment of mitochondrial transport has long been implicated in the pathogenesis of neuropathy and neurodegeneration. However, the role of mitochondria in stabilizing motor nerve terminals at neuromuscular junction (NMJ) remains unclear. We previously demonstrated that mice lacking the antioxidant enzyme, superoxide dismutase-1 (Sod1-/-), develop progressive NMJ denervation. This was rescued by expression of SOD1 exclusively in the mitochondrial intermembrane space (MitoSOD1/Sod1-/-), suggesting that oxidative stress within mitochondria drives denervation in these animals. However, we also observed reduced mitochondrial density in Sod1-/- motor axons in vitro. To investigate the relationship between mitochondrial density and NMJ innervation in vivo, we crossed Sod1-/- mice with the fluorescent reporter strains Thy1-YFP and Thy1-mitoCFP. We identified an age-dependent loss of mitochondria at motor nerve terminals in Sod1-/- mice, that closely correlated with NMJ denervation, and was rescued by MitoSOD1 expression. To test whether augmenting mitochondrial transport rescues Sod1-/- axons, we generated transgenic mice overexpressing the mitochondrial cargo adaptor, Miro1. This led to a partial rescue of mitochondrial density at motor nerve terminals by 12 months of age, but was insufficient to prevent denervation. These findings suggest that loss of mitochondria in the distal motor axon may contribute to denervation in Sod1-/- mice, perhaps via loss of key mitochondrial functions such as calcium buffering and/or energy production.
Collapse
Affiliation(s)
- Lindsey R Hayes
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Seneshaw A Asress
- Department of Neurology and Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Yingjie Li
- Department of Neurology and Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Alexander Galkin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anna Stepanova
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hibiki Kawamata
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jonathan D Glass
- Department of Neurology and Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
56
|
Tapia-Rojas C, Torres AK, Quintanilla RA. Adolescence binge alcohol consumption induces hippocampal mitochondrial impairment that persists during the adulthood. Neuroscience 2019; 406:356-368. [DOI: 10.1016/j.neuroscience.2019.03.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 03/07/2019] [Accepted: 03/09/2019] [Indexed: 01/23/2023]
|
57
|
Calcium Deregulation and Mitochondrial Bioenergetics in GDAP1-Related CMT Disease. Int J Mol Sci 2019; 20:ijms20020403. [PMID: 30669311 PMCID: PMC6359725 DOI: 10.3390/ijms20020403] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/17/2022] Open
Abstract
The pathology of Charcot-Marie-Tooth (CMT), a disease arising from mutations in different genes, has been associated with an impairment of mitochondrial dynamics and axonal biology of mitochondria. Mutations in ganglioside-induced differentiation-associated protein 1 (GDAP1) cause several forms of CMT neuropathy, but the pathogenic mechanisms involved remain unclear. GDAP1 is an outer mitochondrial membrane protein highly expressed in neurons. It has been proposed to play a role in different aspects of mitochondrial physiology, including mitochondrial dynamics, oxidative stress processes, and mitochondrial transport along the axons. Disruption of the mitochondrial network in a neuroblastoma model of GDAP1-related CMT has been shown to decrease Ca2+ entry through the store-operated calcium entry (SOCE), which caused a failure in stimulation of mitochondrial respiration. In this review, we summarize the different functions proposed for GDAP1 and focus on the consequences for Ca2+ homeostasis and mitochondrial energy production linked to CMT disease caused by different GDAP1 mutations.
Collapse
|
58
|
Batllori M, Molero-Luis M, Ormazabal A, Montero R, Sierra C, Ribes A, Montoya J, Ruiz-Pesini E, O'Callaghan M, Pias L, Nascimento A, Palau F, Armstrong J, Yubero D, Ortigoza-Escobar JD, García-Cazorla A, Artuch R. Cerebrospinal fluid monoamines, pterins, and folate in patients with mitochondrial diseases: systematic review and hospital experience. J Inherit Metab Dis 2018; 41:1147-1158. [PMID: 29974349 DOI: 10.1007/s10545-018-0224-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/18/2018] [Accepted: 06/20/2018] [Indexed: 10/28/2022]
Abstract
Mitochondrial diseases are a group of genetic disorders leading to the dysfunction of mitochondrial energy metabolism pathways. We aimed to assess the clinical phenotype and the biochemical cerebrospinal fluid (CSF) biogenic amine profiles of patients with different diagnoses of genetic mitochondrial diseases. We recruited 29 patients with genetically confirmed mitochondrial diseases harboring mutations in either nuclear or mitochondrial DNA (mtDNA) genes. Signs and symptoms of impaired neurotransmission and neuroradiological data were recorded. CSF monoamines, pterins, and 5-methyltetrahydrofolate (5MTHF) concentrations were analyzed using high-performance liquid chromatography with electrochemical and fluorescence detection procedures. The mtDNA mutations were studied by Sanger sequencing, Southern blot, and real-time PCR, and nuclear DNA was assessed either by Sanger or next-generation sequencing. Five out of 29 cases showed predominant dopaminergic signs not attributable to basal ganglia involvement, harboring mutations in different nuclear genes. A chi-square test showed a statistically significant association between high homovanillic acid (HVA) values and low CSF 5-MTHF values (chi-square = 10.916; p = 0.001). Seven out of the eight patients with high CSF HVA values showed cerebral folate deficiency. Five of them harbored mtDNA deletions associated with Kearns-Sayre syndrome (KSS), one had a mitochondrial point mutation at the mtDNA ATPase6 gene, and one had a POLG mutation. In conclusion, dopamine deficiency clinical signs were present in some patients with mitochondrial diseases with different genetic backgrounds. High CSF HVA values, together with a severe cerebral folate deficiency, were observed in KSS patients and in other mtDNA mutation syndromes.
Collapse
Affiliation(s)
- Marta Batllori
- Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Marta Molero-Luis
- Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Aida Ormazabal
- Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Raquel Montero
- Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Sierra
- Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Antonia Ribes
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Institut de Bioquímica Clínica-Corporació Sanitaria Clínic, Barcelona, Spain
| | - Julio Montoya
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Biochemistry, Cellular and Molecular Biology Department, Universidad de Zaragoza, Zaragoza, Spain
| | - Eduardo Ruiz-Pesini
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Biochemistry, Cellular and Molecular Biology Department, Universidad de Zaragoza, Zaragoza, Spain
| | - Mar O'Callaghan
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Pediatric Neurology, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Leticia Pias
- Pediatric Neurology, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Andrés Nascimento
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Pediatric Neurology, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Francesc Palau
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Genetics Department, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Judith Armstrong
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Genetics Department, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Delia Yubero
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Genetics Department, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | | | - Angels García-Cazorla
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
- Pediatric Neurology, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Rafael Artuch
- Clinical Biochemistry, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.
- CIBERER, Instituto de Salud Carlos III, Barcelona, Spain.
- Clinical Biochemistry Department, IRSJD and CIBERER, Hospital Sant Joan de Déu, Passeig Sant Joan de Déu, 2., 08950, Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
59
|
Mitochondrial pathophysiology beyond the retinal ganglion cell: occipital GABA is decreased in autosomal dominant optic neuropathy. Graefes Arch Clin Exp Ophthalmol 2018; 256:2341-2348. [PMID: 30324419 PMCID: PMC6224020 DOI: 10.1007/s00417-018-4153-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/05/2018] [Accepted: 09/24/2018] [Indexed: 01/05/2023] Open
Abstract
PURPOSE It has remained a mystery why some genetic mitochondrial disorders affect predominantly specific cell types such as the retinal ganglion cell. This is particularly intriguing concerning retinal and cortical function since they are tightly linked in health and disease. Autosomal dominant optic neuropathy (ADOA) is a mitochondrial disease that affects the ganglion cell. However, it is unknown whether alterations are also present in the visual cortex, namely in excitation/inhibition balance. METHODS In this study, we performed in vivo structural and biochemical proton magnetic resonance imaging in 14 ADOA and 11 age-matched control participants focusing on the visual cortex, with the aim of establishing whether in this genetically determined disease an independent cortical neurochemical phenotype could be established irrespective of a putative structural phenotype. Cortical thickness of anatomically defined visual areas was estimated, and a voxel-based morphometry approach was used to assess occipital volumetric changes in ADOA. Neurochemical measurements were focused on γ-aminobutyric acid (GABA) and glutamate, as indicators of the local excitatory/inhibitory balance. RESULTS We found evidence for reduced visual cortical GABA and preserved glutamate concentrations in the absence of cortical or subcortical atrophy. These changes in GABA levels were explained by neither structural nor functional measures of visual loss, suggesting a developmental origin. CONCLUSIONS These results suggest that mitochondrial disorders that were previously believed to only affect retinal function may also affect cortical physiology, especially the GABAergic system, suggesting reduced brain inhibition vs. excitation. This GABA phenotype, independent of sensory loss or cortical atrophy and in the presence of preserved glutamate levels, suggests a neurochemical developmental change at the cortical level, leading to a pathophysiological excitation/inhibition imbalance.
Collapse
|
60
|
Kumar A, Foster TC. Alteration in NMDA Receptor Mediated Glutamatergic Neurotransmission in the Hippocampus During Senescence. Neurochem Res 2018; 44:38-48. [PMID: 30209673 DOI: 10.1007/s11064-018-2634-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/07/2018] [Accepted: 09/08/2018] [Indexed: 12/17/2022]
Abstract
Glutamate is the primary excitatory neurotransmitter in neurons and glia. N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and kainate receptors are major ionotropic glutamate receptors. Glutamatergic neurotransmission is strongly linked with Ca2+ homeostasis. Research has provided ample evidence that brain aging is associated with altered glutamatergic neurotransmission and Ca2+ dysregulation. Much of the work has focused on the hippocampus, a brain region critically involved in learning and memory, which is particularly susceptible to dysfunction during senescence. The current review examines Ca2+ regulation with a focus on the NMDA receptors in the hippocampus. Integrating the knowledge of the complexity of age-related alterations in Ca2+ homeostasis and NMDA receptor-mediated glutamatergic neurotransmission will positively shape the development of highly effective therapeutics to treat brain disorders including cognitive impairment.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL, 32610-0244, USA.
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL, 32610-0244, USA.
- Genetics and Genomics Program, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
61
|
Bellesi M, de Vivo L, Koebe S, Tononi G, Cirelli C. Sleep and Wake Affect Glycogen Content and Turnover at Perisynaptic Astrocytic Processes. Front Cell Neurosci 2018; 12:308. [PMID: 30254569 PMCID: PMC6141665 DOI: 10.3389/fncel.2018.00308] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/23/2018] [Indexed: 12/26/2022] Open
Abstract
Astrocytic glycogen represents the only form of glucose storage in the brain, and one of the outcomes of its breakdown is the production of lactate that can be used by neurons as an alternative energetic substrate. Since brain metabolism is higher in wake than in sleep, it was hypothesized that glycogen stores are depleted during wake and replenished during sleep. Furthermore, it was proposed that glycogen depletion leads to the progressive increase in adenosine levels during wake, providing a homeostatic signal that reflects the buildup of sleep pressure. However, previous studies that measured glycogen dynamics across the sleep/wake cycle obtained inconsistent results, and only measured glycogen in whole tissue. Since most energy in the brain is used to sustain synaptic activity, here we employed tridimensional electron microscopy to quantify glycogen content in the astrocytic processes surrounding the synapse. We studied axon-spine synapses in the frontal cortex of young mice after ~7 h of sleep, 7–8 h of spontaneous or forced wake, or 4.5 days of sleep restriction. Relative to sleep, all wake conditions increased the number of glycogen granules around the synapses to a similar extent. However, progressively longer periods of wake were associated with progressively smaller glycogen granules, suggesting increased turnover. Despite the increased number of granules, in all wake conditions the estimated amount of glucose within the granules was lower than in sleep, indicating that sleep may favor glucose storage. Finally, chronic sleep restriction moved glycogen granules closer to the synaptic cleft. Thus, both short and long wake lead to increased glycogen turnover around cortical synapses, whereas sleep promotes glycogen accumulation.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Samuel Koebe
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
62
|
Jara C, Aránguiz A, Cerpa W, Tapia-Rojas C, Quintanilla RA. Genetic ablation of tau improves mitochondrial function and cognitive abilities in the hippocampus. Redox Biol 2018; 18:279-294. [PMID: 30077079 PMCID: PMC6072970 DOI: 10.1016/j.redox.2018.07.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/13/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022] Open
Abstract
Tau is a key protein for microtubule stability; however, post-translationally modified tau contributes to neurodegenerative diseases by forming tau aggregates in the neurons. Previous reports from our group and others have shown that pathological forms of tau are toxic and impair mitochondrial function, whereas tau deletion is neuroprotective. However, the effects of tau ablation on brain structure and function in young mice have not been fully elucidated. Therefore, the aim of this study was to investigate the implications of tau ablation on the mitochondrial function and cognitive abilities of a litter of young mice (3 months old). Our results showed that tau deletion had positive effects on hippocampal cells by decreasing oxidative damage, favoring a mitochondrial pro-fusion state, and inhibiting mitochondrial permeability transition pore (mPTP) formation by reducing cyclophilin D (Cyp-D) protein. More importantly, tau deletion increased ATP production and improved the recognition memory and attentive capacity of juvenile mice. Therefore, the absence of tau enhanced brain function by improving mitochondrial health, which supplied more energy to the synapses. Thus, our work opens the possibility that preventing negative tau modifications could enhance brain function through the improvement of mitochondrial health.
Collapse
Affiliation(s)
- Claudia Jara
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Chile
| | - Alejandra Aránguiz
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Chile.
| | | |
Collapse
|
63
|
Hill RL, Kulbe JR, Singh IN, Wang JA, Hall ED. Synaptic Mitochondria are More Susceptible to Traumatic Brain Injury-induced Oxidative Damage and Respiratory Dysfunction than Non-synaptic Mitochondria. Neuroscience 2018; 386:265-283. [PMID: 29960045 DOI: 10.1016/j.neuroscience.2018.06.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/12/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) results in mitochondrial dysfunction and induction of lipid peroxidation (LP). Lipid peroxidation-derived neurotoxic aldehydes such as 4-HNE and acrolein bind to mitochondrial proteins, inducing additional oxidative damage and further exacerbating mitochondrial dysfunction and LP. Mitochondria are heterogeneous, consisting of both synaptic and non-synaptic populations. Synaptic mitochondria are reported to be more vulnerable to injury; however, this is the first study to characterize the temporal profile of synaptic and non-synaptic mitochondria following TBI, including investigation of respiratory dysfunction and oxidative damage to mitochondrial proteins between 3 and 120 h following injury. These results indicate that synaptic mitochondria are indeed the more vulnerable population, showing both more rapid and severe impairments than non-synaptic mitochondria. By 24 h, synaptic respiration is significantly impaired compared to synaptic sham, whereas non-synaptic respiration does not decline significantly until 48 h. Decreases in respiration are associated with increases in oxidative damage to synaptic and non-synaptic mitochondrial proteins at 48 h and 72 h, respectively. These results indicate that the therapeutic window for mitochondria-targeted pharmacological neuroprotectants to prevent respiratory dysfunction is shorter for the more vulnerable synaptic mitochondria than for the non-synaptic population.
Collapse
Affiliation(s)
- Rachel L Hill
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Jacqueline R Kulbe
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Indrapal N Singh
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Juan A Wang
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Edward D Hall
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States.
| |
Collapse
|
64
|
Abstract
Even the simplest animals possess sophisticated systems for sensing and securing nutrients. After all, ensuring adequate nutrition is essential for sustaining life. Once multicellular animals grew too large to be nourished by simple diffusion of nutrients from their environment, they required a digestive system for the absorption and digestion of food. The majority of cells in the digestive tract are enterocytes that are designed to absorb nutrients. However, the digestive tracts of animals ranging from worms to humans contain specialized cells that discriminate between nutrients and nondigestible ingestants. These cells "sense" both the environment within the gut lumen and nutrients as they cross the gut epithelium. This dual sensing is then translated into local signals that regulate the gut epithelium or distant signals through hormones or nerves. This review will discuss how sensors of the gut interact with cells of the epithelium and neurons to regulate epithelial integrity and initiate neural transmission from the gut lumen. © 2017 American Physiological Society. Compr Physiol 8:1019-1030, 2018.
Collapse
Affiliation(s)
- Rodger A Liddle
- Department of Medicine, Duke University and Durham VA Healthcare System, Durham, North Carolina, USA
| |
Collapse
|
65
|
Nesterov SV, Skorobogatova YA, Panteleeva AA, Pavlik LL, Mikheeva IB, Yaguzhinsky LS, Nartsissov YR. NMDA and GABA receptor presence in rat heart mitochondria. Chem Biol Interact 2018; 291:40-46. [PMID: 29883723 DOI: 10.1016/j.cbi.2018.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/30/2018] [Accepted: 06/05/2018] [Indexed: 11/19/2022]
Abstract
The purpose of this study is to demonstrate the presence of three more receptors in mitochondria. Two N-methyl-d-aspartate receptor (NMDAR) subunits (NR1 and NR2B) are found by protein immunoblot and immunogold labeling in mitochondria fraction isolated from rat heart. These data allow supposing NMDAR presence and functioning in the inner mitochondrial membrane. There are no signs of receptor presence obtained in heart tissue lysate, that indicates the receptor localization exactly in mitochondria. The possible receptor functions discussed are its participation in calcium transport and in excitation-metabolism coupling. Besides, preliminary evidence is obtained of GABAA and GABAB receptors presence in heart mitochondria. One can surmise their role in metabolism regulation and their possible co-operation with NMDAR just as in the nervous system.
Collapse
Affiliation(s)
- Semen V Nesterov
- Moscow Institute of Physics and Technology, Institutskiy pereulok, 9, Dolgoprudny, 141700, Russia.
| | - Yulia A Skorobogatova
- Moscow Institute of Physics and Technology, Institutskiy pereulok, 9, Dolgoprudny, 141700, Russia.
| | - Alisa A Panteleeva
- Belozersky Research Institute for Physico Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| | - Lyubov L Pavlik
- Institute of Theoretical and Experimental Biophisics Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Irina B Mikheeva
- Institute of Theoretical and Experimental Biophisics Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Lev S Yaguzhinsky
- Institute of Cytochemistry and Molecular Pharmacology, 6-th Radialnaya str. 24-14, Moscow, 115404, Russia; Belozersky Research Institute for Physico Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| | - Yaroslav R Nartsissov
- Institute of Cytochemistry and Molecular Pharmacology, 6-th Radialnaya str. 24-14, Moscow, 115404, Russia.
| |
Collapse
|
66
|
Cuperfain AB, Zhang ZL, Kennedy JL, Gonçalves VF. The Complex Interaction of Mitochondrial Genetics and Mitochondrial Pathways in Psychiatric Disease. MOLECULAR NEUROPSYCHIATRY 2018; 4:52-69. [PMID: 29998118 DOI: 10.1159/000488031] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/27/2018] [Indexed: 12/18/2022]
Abstract
While accounting for only 2% of the body's weight, the brain utilizes up to 20% of the body's total energy. Not surprisingly, metabolic dysfunction and energy supply-and-demand mismatch have been implicated in a variety of neurological and psychiatric disorders. Mitochondria are responsible for providing the brain with most of its energetic demands, and the brain uses glucose as its exclusive energy source. Exploring the role of mitochondrial dysfunction in the etiology of psychiatric disease is a promising avenue to investigate further. Genetic analysis of mitochondrial activity is a cornerstone in understanding disease pathogenesis related to metabolic dysfunction. In concert with neuroimaging and pathological study, genetics provides an important bridge between biochemical findings and clinical correlates in psychiatric disease. Mitochondrial genetics has several unique aspects to its analysis, and corresponding special considerations. Here, we review the components of mitochondrial genetic analysis - nuclear DNA, mitochon-drial DNA, mitochondrial pathways, pseudogenes, nuclear-mitochondrial mismatch, and microRNAs - that could contribute to an observable clinical phenotype. Throughout, we highlight psychiatric diseases that can arise due to dysfunction in these processes, with a focus on schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Ari B Cuperfain
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Zhi Lun Zhang
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - James L Kennedy
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Vanessa F Gonçalves
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
67
|
Gonçalves VF, Cappi C, Hagen CM, Sequeira A, Vawter MP, Derkach A, Zai CC, Hedley PL, Bybjerg-Grauholm J, Pouget JG, Cuperfain AB, Sullivan PF, Christiansen M, Kennedy JL, Sun L. A Comprehensive Analysis of Nuclear-Encoded Mitochondrial Genes in Schizophrenia. Biol Psychiatry 2018; 83:780-789. [PMID: 29628042 PMCID: PMC7168759 DOI: 10.1016/j.biopsych.2018.02.1175] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND The genetic risk factors of schizophrenia (SCZ), a severe psychiatric disorder, are not yet fully understood. Multiple lines of evidence suggest that mitochondrial dysfunction may play a role in SCZ, but comprehensive association studies are lacking. We hypothesized that variants in nuclear-encoded mitochondrial genes influence susceptibility to SCZ. METHODS We conducted gene-based and gene-set analyses using summary association results from the Psychiatric Genomics Consortium Schizophrenia Phase 2 (PGC-SCZ2) genome-wide association study comprising 35,476 cases and 46,839 control subjects. We applied the MAGMA method to three sets of nuclear-encoded mitochondrial genes: oxidative phosphorylation genes, other nuclear-encoded mitochondrial genes, and genes involved in nucleus-mitochondria crosstalk. Furthermore, we conducted a replication study using the iPSYCH SCZ sample of 2290 cases and 21,621 control subjects. RESULTS In the PGC-SCZ2 sample, 1186 mitochondrial genes were analyzed, among which 159 had p values < .05 and 19 remained significant after multiple testing correction. A meta-analysis of 818 genes combining the PGC-SCZ2 and iPSYCH samples resulted in 104 nominally significant and nine significant genes, suggesting a polygenic model for the nuclear-encoded mitochondrial genes. Gene-set analysis, however, did not show significant results. In an in silico protein-protein interaction network analysis, 14 mitochondrial genes interacted directly with 158 SCZ risk genes identified in PGC-SCZ2 (permutation p = .02), and aldosterone signaling in epithelial cells and mitochondrial dysfunction pathways appeared to be overrepresented in this network of mitochondrial and SCZ risk genes. CONCLUSIONS This study provides evidence that specific aspects of mitochondrial function may play a role in SCZ, but we did not observe its broad involvement even using a large sample.
Collapse
Affiliation(s)
- Vanessa F Gonçalves
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
| | - Carolina Cappi
- Department of Psychiatry, University of São Paulo, School of Medicine, Brazil
| | - Christian M Hagen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Adolfo Sequeira
- Department of Psychiatry and Human Behavior, University of California, Irvine, USA
| | - Marquis P Vawter
- Department of Psychiatry and Human Behavior, University of California, Irvine, USA
| | - Andriy Derkach
- Department of Statistical Sciences, Faculty of Arts and Science, University of Toronto, Toronto, Canada
| | - Clement C Zai
- Department of Psychiatry, University of Toronto, Toronto, Canada,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Canada
| | - Paula L Hedley
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | | | - Jennie G Pouget
- Department of Psychiatry, University of Toronto, Toronto, Canada,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Canada
| | - Ari B. Cuperfain
- Department of Psychiatry, University of Toronto, Toronto, Canada,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Canada
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA;,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark,Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| | - James L Kennedy
- Department of Psychiatry, University of Toronto, Toronto, Canada,Neuroscience Section, Centre for Addiction and Mental Health, Toronto, Canada,Corresponding author: Vanessa F Gonçalves ()
| | - Lei Sun
- Department of Statistical Sciences, Faculty of Arts and Science, University of Toronto, Toronto, Canada,Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada,Corresponding author: Vanessa F Gonçalves ()
| |
Collapse
|
68
|
Wang E, Zhu H, Wang X, Gower AC, Wallack M, Blusztajn JK, Kowall N, Qiu WQ. Amylin Treatment Reduces Neuroinflammation and Ameliorates Abnormal Patterns of Gene Expression in the Cerebral Cortex of an Alzheimer's Disease Mouse Model. J Alzheimers Dis 2018; 56:47-61. [PMID: 27911303 DOI: 10.3233/jad-160677] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our recent study has demonstrated that peripheral amylin treatment reduces the amyloid pathology in the brain of Alzheimer's disease (AD) mouse models, and improves their learning and memory. We hypothesized that the beneficial effects of amylin for AD was beyond reducing the amyloids in the brain, and have now directly tested the actions of amylin on other aspects of AD pathogenesis, especially neuroinflammation. A 10-week course of peripheral amylin treatment significantly reduced levels of cerebral inflammation markers, Cd68 and Iba1, in amyloid precursor protein (APP) transgenic mice. Mechanistic studies indicated the protective effect of amylin required interaction with its cognate receptor because silencing the amylin receptor expression blocked the amylin effect on Cd68 in microglia. Using weighted gene co-expression network analysis, we discovered that amylin treatment influenced two gene modules linked with amyloid pathology: 1) a module related to proinflammation and transport/vesicle process that included a hub gene of Cd68, and 2) a module related to mitochondria function that included a hub gene of Atp5b. Amylin treatment restored the expression of most genes in the APP cortex toward levels observed in the wild-type (WT) cortex in these two modules including Cd68 and Atp5b. Using a human dataset, we found that the expression levels of Cd68 and Atp5b were significantly correlated with the neurofibrillary tangle burden in the AD brain and with their cognition. These data suggest that amylin acts on the pathological cascade in animal models of AD, and further supports the therapeutic potential of amylin-type peptides for AD.
Collapse
Affiliation(s)
- Erming Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Xiaofan Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University School of Medicine, Boston, MA, USA
| | - Max Wallack
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Jan Krzysztof Blusztajn
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Neil Kowall
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA.,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
69
|
Doktór B, Damulewicz M, Krzeptowski W, Bednarczyk B, Pyza E. Effects of PINK1 mutation on synapses and behavior in the brain of Drosophila melanogaster. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
70
|
Rittschof CC, Vekaria HJ, Palmer JH, Sullivan PG. Brain mitochondrial bioenergetics change with rapid and prolonged shifts in aggression in the honey bee, Apis mellifera. J Exp Biol 2018; 221:jeb.176917. [DOI: 10.1242/jeb.176917] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022]
Abstract
Neuronal function demands high-level energy production, and as such, a decline in mitochondrial respiration characterizes brain injury and disease. A growing number of studies, however, link brain mitochondrial function to behavioral modulation in non-diseased contexts. In the honey bee, we show for the first time that an acute social interaction, which invokes an aggressive response, may also cause a rapid decline in brain mitochondrial bioenergetics. The degree and speed of this decline has only been previously observed in the context of brain injury. Furthermore, in the honey bee, age-related increases in aggressive tendency are associated with increased baseline brain mitochondrial respiration, as well as increased plasticity in response to metabolic fuel type in vitro. Similarly, diet restriction and ketone body feeding, which commonly enhance mammalian brain mitochondrial function in vivo, cause increased aggression. Thus, even in normal behavioral contexts, brain mitochondria show a surprising degree of variation in function over both rapid and prolonged timescales, with age predicting both baseline function and plasticity in function. These results suggest that mitochondrial function is integral to modulating aggression-related neuronal signaling. We hypothesize that variation in function reflects mitochondrial calcium buffering activity, and that shifts in mitochondrial function signal to the neuronal soma to regulate gene expression and neural energetic state. Modulating brain energetic state is emerging as a critical component of the regulation of behavior in non-diseased contexts.
Collapse
Affiliation(s)
- Clare C. Rittschof
- Department of Entomology, University of Kentucky, S-225 Ag. Science Center North, Lexington, KY, 40546, USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center and the Department of Neuroscience, University of Kentucky, 741 South Limestone Street, 475 BBSRB, Lexington, KY 40536-0509, USA
| | - Joseph H. Palmer
- Department of Entomology, University of Kentucky, S-225 Ag. Science Center North, Lexington, KY, 40546, USA
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center and the Department of Neuroscience, University of Kentucky, 741 South Limestone Street, 475 BBSRB, Lexington, KY 40536-0509, USA
| |
Collapse
|
71
|
Ahmad F, Salahuddin M, Alamoudi W, Acharya S. Dysfunction of cortical synapse-specific mitochondria in developing rats exposed to lead and its amelioration by ascorbate supplementation. Neuropsychiatr Dis Treat 2018; 14:813-824. [PMID: 29606875 PMCID: PMC5868605 DOI: 10.2147/ndt.s148248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Lead (Pb) is a widespread environmental neurotoxin and its exposure even in minute quantities can lead to compromised neuronal functions. A developing brain is particularly vulnerable to Pb mediated toxicity and early-life exposure leads to permanent alterations in brain development and neuronal signaling and plasticity, culminating into cognitive and behavioral dysfunctions and elevated risk of neuropsychiatric disorders later in life. Nevertheless, the underlying biochemical mechanisms have not been completely discerned. METHODS Because of their ability to fulfill high energy needs and to act as calcium buffers in events of high intensity neuronal activity as well as their adaptive regulatory capability to match the requirements of the dynamicity of synaptic signaling, synapse-specific or synaptic mitochondria (SM) are critical for synaptic development, function and plasticity. Our aim for the present study hence was to characterize the effects of early-life Pb exposure on the functions of SM of prepubertal rats. For this purpose, employing a chronic model of Pb neurotoxicity, we exposed rat pups perinatally and postnatally to Pb and used a plethora of colorimetric and fluorometric assays for assessing redox and bioenergetic properties of SM. In addition, taking advantage of its ability as an antioxidant and as a metal chelator, we employed ascorbic acid (vitamin C) supplementation as an ameliorative therapeutic strategy against Pb-induced neurotoxicity and dysfunction of SM. RESULTS Our results suggest that early-life exposure to Pb leads to elevated oxidative stress in cortical SM with consequent compromises in its energy metabolism activity. Ascorbate supplementation resulted in significant recovery of Pb-induced oxidative stress and functional compromise of SM. CONCLUSION Alterations in redox status and bioenergetic properties of SM could potentially contribute to the synaptic dysfunction observed in events of Pb neurotoxicity. Additionally, our study provides evidence for suitability of ascorbate as a significant ameliorative agent in tacking Pb neurotoxicity.
Collapse
Affiliation(s)
- Faraz Ahmad
- Department of Public Health, College of Public Health, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Neuroscience Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammad Salahuddin
- Animal House Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Widyan Alamoudi
- Neuroscience Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sadananda Acharya
- Department of Public Health, College of Public Health, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
72
|
Tapia-Rojas C, Mira RG, Torres AK, Jara C, Pérez MJ, Vergara EH, Cerpa W, Quintanilla RA. Alcohol consumption during adolescence: A link between mitochondrial damage and ethanol brain intoxication. Birth Defects Res 2017; 109:1623-1639. [DOI: 10.1002/bdr2.1172] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/31/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Cheril Tapia-Rojas
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Rodrigo G. Mira
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago 8331150 Chile
| | - Angie K. Torres
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Claudia Jara
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - María José Pérez
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Erick H. Vergara
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| | - Waldo Cerpa
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas; Pontificia Universidad Católica de Chile; Santiago 8331150 Chile
| | - Rodrigo A. Quintanilla
- Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA); Santiago Chile
- Laboratory of Neurodegenerative Diseases; Universidad Autónoma de Chile; Chile
| |
Collapse
|
73
|
Graham LC, Eaton SL, Brunton PJ, Atrih A, Smith C, Lamont DJ, Gillingwater TH, Pennetta G, Skehel P, Wishart TM. Proteomic profiling of neuronal mitochondria reveals modulators of synaptic architecture. Mol Neurodegener 2017; 12:77. [PMID: 29078798 PMCID: PMC5659037 DOI: 10.1186/s13024-017-0221-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 10/19/2017] [Indexed: 02/16/2023] Open
Abstract
Background Neurons are highly polarized cells consisting of three distinct functional domains: the cell body (and associated dendrites), the axon and the synapse. Previously, it was believed that the clinical phenotypes of neurodegenerative diseases were caused by the loss of entire neurons, however it has recently become apparent that these neuronal sub-compartments can degenerate independently, with synapses being particularly vulnerable to a broad range of stimuli. Whilst the properties governing the differential degenerative mechanisms remain unknown, mitochondria consistently appear in the literature, suggesting these somewhat promiscuous organelles may play a role in affecting synaptic stability. Synaptic and non-synaptic mitochondrial subpools are known to have different enzymatic properties (first demonstrated by Lai et al., 1977). However, the molecular basis underpinning these alterations, and their effects on morphology, has not been well documented. Methods The current study has employed electron microscopy, label-free proteomics and in silico analyses to characterize the morphological and biochemical properties of discrete sub-populations of mitochondria. The physiological relevance of these findings was confirmed in-vivo using a molecular genetic approach at the Drosophila neuromuscular junction. Results Here, we demonstrate that mitochondria at the synaptic terminal are indeed morphologically different to non-synaptic mitochondria, in both rodents and human patients. Furthermore, generation of proteomic profiles reveals distinct molecular fingerprints – highlighting that the properties of complex I may represent an important specialisation of synaptic mitochondria. Evidence also suggests that at least 30% of the mitochondrial enzymatic activity differences previously reported can be accounted for by protein abundance. Finally, we demonstrate that the molecular differences between discrete mitochondrial sub-populations are capable of selectively influencing synaptic morphology in-vivo. We offer several novel mitochondrial candidates that have the propensity to significantly alter the synaptic architecture in-vivo. Conclusions Our study demonstrates discrete proteomic profiles exist dependent upon mitochondrial subcellular localization and selective alteration of intrinsic mitochondrial proteins alters synaptic morphology in-vivo. Electronic supplementary material The online version of this article (10.1186/s13024-017-0221-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura C Graham
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Samantha L Eaton
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Paula J Brunton
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Abdelmadjid Atrih
- FingerPrints Proteomics Facility, College of Life Sciences, University of Dundee, Dundee, UK
| | - Colin Smith
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Department of Academic Neuropathology, University of Edinburgh, CCBS, Chancellor's Building, Little France, Edinburgh, UK
| | - Douglas J Lamont
- FingerPrints Proteomics Facility, College of Life Sciences, University of Dundee, Dundee, UK
| | - Thomas H Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - Giuseppa Pennetta
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - Paul Skehel
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - Thomas M Wishart
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK. .,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
74
|
Djeungoue-Petga MA, Hebert-Chatelain E. Linking Mitochondria and Synaptic Transmission: The CB1 Receptor. Bioessays 2017; 39. [PMID: 29058339 DOI: 10.1002/bies.201700126] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/19/2017] [Indexed: 12/21/2022]
Abstract
CB1 receptors are functionally present within brain mitochondria (mtCB1), although they are usually considered specifically targeted to plasma membrane. Acute activation of mtCB1 alters mitochondrial ATP generation, synaptic transmission, and memory performance. However, the detailed mechanism linking disrupted mitochondrial metabolism and synaptic transmission is still uncharacterized. CB1 receptors are among the most abundant G protein-coupled receptors in the brain and impact on several processes, including fear coping, anxiety, stress, learning, and memory. Mitochondria perform several key physiological processes for neuronal homeostasis, including production of ATP and reactive oxygen species, calcium buffering, metabolism of neurotransmitters, and apoptosis. It is therefore possible that acute activation of mtCB1 impacts on these different mitochondrial functions to modulate synaptic transmission. In reviewing and integrating across the literature in this area, we describe the possible mechanisms involved in the regulation of brain physiology by mtCB1 receptors.
Collapse
Affiliation(s)
| | - Etienne Hebert-Chatelain
- Department of Biology, Université de Moncton, 18 Av Antonine Maillet, Moncton, New Brunswick, Canada
| |
Collapse
|
75
|
Ben-Shachar D. Mitochondrial multifaceted dysfunction in schizophrenia; complex I as a possible pathological target. Schizophr Res 2017; 187:3-10. [PMID: 27802911 DOI: 10.1016/j.schres.2016.10.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/10/2016] [Accepted: 10/14/2016] [Indexed: 01/09/2023]
Abstract
Mitochondria are key players in various essential cellular processes beyond being the main energy supplier of the cell. Accordingly, they are involved in neuronal synaptic transmission, neuronal growth and sprouting and consequently neuronal plasticity and connectivity. In addition, mitochondria participate in the modulation of gene transcription and inflammation as well in physiological responses in health and disease. Schizophrenia is currently regarded as a neurodevelopmental disorder associated with impaired immune system, aberrant neuronal differentiation and abnormalities in various neurotransmitter systems mainly the dopaminergic, glutaminergic and GABAergic. Ample evidence has been accumulated over the last decade indicating a multifaceted dysfunction of mitochondria in schizophrenia. Indeed, mitochondrial deficit can be of relevance for the majority of the pathologies observed in this disease. In the present article, we overview specific deficits of the mitochondria in schizophrenia, with a focus on the first complex (complex I) of the mitochondrial electron transport chain (ETC). We argue that complex I, being a major factor in the regulation of mitochondrial ETC, is a possible key modulator of various functions of the mitochondria. We review biochemical, molecular, cellular and functional evidence for mitochondrial impairments and their possible convergence to impact in-vitro neuronal differentiation efficiency in schizophrenia. Mitochondrial function in schizophrenia may advance our knowledge of the disease pathophysiology and open the road for new treatment targets for the benefit of the patients.
Collapse
Affiliation(s)
- Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion-IIT, Haifa, Israel.
| |
Collapse
|
76
|
Bryan K, McGivney BA, Farries G, McGettigan PA, McGivney CL, Gough KF, MacHugh DE, Katz LM, Hill EW. Equine skeletal muscle adaptations to exercise and training: evidence of differential regulation of autophagosomal and mitochondrial components. BMC Genomics 2017; 18:595. [PMID: 28793853 PMCID: PMC5551008 DOI: 10.1186/s12864-017-4007-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 08/02/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND A single bout of exercise induces changes in gene expression in skeletal muscle. Regular exercise results in an adaptive response involving changes in muscle architecture and biochemistry, and is an effective way to manage and prevent common human diseases such as obesity, cardiovascular disorders and type II diabetes. However, the biomolecular mechanisms underlying such responses still need to be fully elucidated. Here we performed a transcriptome-wide analysis of skeletal muscle tissue in a large cohort of untrained Thoroughbred horses (n = 51) before and after a bout of high-intensity exercise and again after an extended period of training. We hypothesized that regular high-intensity exercise training primes the transcriptome for the demands of high-intensity exercise. RESULTS An extensive set of genes was observed to be significantly differentially regulated in response to a single bout of high-intensity exercise in the untrained cohort (3241 genes) and following multiple bouts of high-intensity exercise training over a six-month period (3405 genes). Approximately one-third of these genes (1025) and several biological processes related to energy metabolism were common to both the exercise and training responses. We then developed a novel network-based computational analysis pipeline to test the hypothesis that these transcriptional changes also influence the contextual molecular interactome and its dynamics in response to exercise and training. The contextual network analysis identified several important hub genes, including the autophagosomal-related gene GABARAPL1, and dynamic functional modules, including those enriched for mitochondrial respiratory chain complexes I and V, that were differentially regulated and had their putative interactions 're-wired' in the exercise and/or training responses. CONCLUSION Here we have generated for the first time, a comprehensive set of genes that are differentially expressed in Thoroughbred skeletal muscle in response to both exercise and training. These data indicate that consecutive bouts of high-intensity exercise result in a priming of the skeletal muscle transcriptome for the demands of the next exercise bout. Furthermore, this may also lead to an extensive 're-wiring' of the molecular interactome in both exercise and training and include key genes and functional modules related to autophagy and the mitochondrion.
Collapse
Affiliation(s)
- Kenneth Bryan
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Beatrice A. McGivney
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Gabriella Farries
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Paul A. McGettigan
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Charlotte L. McGivney
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Katie F. Gough
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| | - David E. MacHugh
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Lisa M. Katz
- UCD School of Veterinary Medicine, University College Dublin, Belfield, D04 V1W8 Ireland
| | - Emmeline W. Hill
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Ireland
| |
Collapse
|
77
|
Duka T, Collins Z, Anderson SM, Raghanti MA, Ely JJ, Hof PR, Wildman DE, Goodman M, Grossman LI, Sherwood CC. Divergent lactate dehydrogenase isoenzyme profile in cellular compartments of primate forebrain structures. Mol Cell Neurosci 2017; 82:137-142. [PMID: 28461219 PMCID: PMC5531073 DOI: 10.1016/j.mcn.2017.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 04/13/2017] [Accepted: 04/20/2017] [Indexed: 10/19/2022] Open
Abstract
The compartmentalization and association of lactate dehydrogenase (LDH) with specific cellular structures (e.g., synaptosomal, sarcoplasmic or mitochondrial) may play an important role in brain energy metabolism. Our previous research revealed that LDH in the synaptosomal fraction shifts toward the aerobic isoforms (LDH-B) among the large-brained haplorhine primates compared to strepsirrhines. Here, we further analyzed the subcellular localization of LDH in primate forebrain structures using quantitative Western blotting and ELISA. We show that, in cytosolic and mitochondrial subfractions, LDH-B expression level was relatively elevated and LDH-A declined in haplorhines compared to strepsirrhines. LDH-B expression in mitochondrial fractions of the neocortex was preferentially increased, showing a particularly significant rise in the ratio of LDH-B to LDH-A in chimpanzees and humans. We also found a significant correlation between the protein levels of LDH-B in mitochondrial fractions from haplorhine neocortex and the synaptosomal LDH-B that suggests LDH isoforms shift from a predominance of A-subunits toward B-subunits as part of a system that spatially buffers dynamic energy requirements of brain cells. Our results indicate that there is differential subcellular compartmentalization of LDH isoenzymes that evolved among different primate lineages to meet the energy requirements in neocortical and striatal cells.
Collapse
Affiliation(s)
- Tetyana Duka
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA
| | - Zachary Collins
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA
| | - Sarah M Anderson
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | | | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Derek E Wildman
- Department of Molecular and Integrative Physiology and Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Morris Goodman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
78
|
Pérez MJ, Vergara-Pulgar K, Jara C, Cabezas-Opazo F, Quintanilla RA. Caspase-Cleaved Tau Impairs Mitochondrial Dynamics in Alzheimer’s Disease. Mol Neurobiol 2017; 55:1004-1018. [DOI: 10.1007/s12035-017-0385-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/04/2017] [Indexed: 11/29/2022]
|
79
|
Pereira JC, Pradella Hallinan M, Alves RC. Secondary to excessive melatonin synthesis, the consumption of tryptophan from outside the blood-brain barrier and melatonin over-signaling in the pars tuberalis may be central to the pathophysiology of winter depression. Med Hypotheses 2017; 98:69-75. [DOI: 10.1016/j.mehy.2016.11.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/27/2016] [Indexed: 12/17/2022]
|
80
|
Modeling Axonal Defects in Hereditary Spastic Paraplegia with Human Pluripotent Stem Cells. ACTA ACUST UNITED AC 2016; 11:339-354. [PMID: 27956894 DOI: 10.1007/s11515-016-1416-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cortical motor neurons, also known as upper motor neurons, are large projection neurons whose axons convey signals to lower motor neurons to control the muscle movements. Degeneration of cortical motor neuron axons is implicated in several debilitating disorders, including hereditary spastic paraplegia (HSP) and amyotrophic lateral sclerosis (ALS). Since the discovery of the first HSP gene, SPAST that encodes spastin, over 70 distinct genetic loci associated with HSP have been identified. How the mutations of these functionally diverse genes result in axonal degeneration and why certain axons are affected in HSP remains largely unknown. The development of induced pluripotent stem cell (iPSC) technology has provided researchers an excellent resource to generate patient-specific human neurons to model human neuropathologic processes including axonal defects. METHODS In this article, we will frst review the pathology and pathways affected in the common forms of HSP subtypes by searching the PubMed database. We will then summurize the findings and insights gained from studies using iPSC-based models, and discuss the challenges and future directions. RESULTS HSPs, a heterogeneous group of genetic neurodegenerative disorders, are characterized by lower extremity weakness and spasticity that result from retrograde axonal degeneration of cortical motor neurons. Recently, iPSCs have been generated from several common forms of HSP including SPG4, SPG3A, and SPG11 patients. Neurons derived from HSP iPSCs exhibit disease-relevant axonal defects, such as impaired neurite outgrowth, increased axonal swellings, and reduced axonal transport. CONCLUSION These patient-derived neurons offer unique tools to study the pathogenic mechanisms and explore the treatments for rescuing axonal defects in HSP, as well as other diseases involving axonopathy.
Collapse
|
81
|
Kwon SK, Hirabayashi Y, Polleux F. Organelle-Specific Sensors for Monitoring Ca 2+ Dynamics in Neurons. Front Synaptic Neurosci 2016; 8:29. [PMID: 27695411 PMCID: PMC5025517 DOI: 10.3389/fnsyn.2016.00029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/30/2016] [Indexed: 11/16/2022] Open
Abstract
Calcium (Ca2+) plays innumerable critical functions in neurons ranging from regulation of neurotransmitter release and synaptic plasticity to activity-dependent transcription. Therefore, more than any other cell types, neurons are critically dependent on spatially and temporally controlled Ca2+ dynamics. This is achieved through an exquisite level of compartmentalization of Ca2+ storage and release from various organelles. The function of these organelles in the regulation of Ca2+ dynamics has been studied for decades using electrophysiological and optical methods combined with pharmacological and genetic alterations. Mitochondria and the endoplasmic reticulum (ER) are among the organelles playing the most critical roles in Ca2+ dynamics in neurons. At presynaptic boutons, Ca2+ triggers neurotransmitter release and synaptic plasticity, and postsynaptically, Ca2+ mobilization mediates long-term synaptic plasticity. To explore Ca2+ dynamics in live cells and intact animals, various synthetic and genetically encoded fluorescent Ca2+ sensors were developed, and recently, many groups actively increased the sensitivity and diversity of genetically encoded Ca2+ indicators (GECIs). Following conjugation with various signal peptides, these improved GECIs can be targeted to specific subcellular compartments, allowing monitoring of organelle-specific Ca2+ dynamics. Here, we review recent findings unraveling novel roles for mitochondria- and ER-dependent Ca2+ dynamics in neurons and at synapses.
Collapse
Affiliation(s)
- Seok-Kyu Kwon
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Kavli Institute for Brain Science, Columbia University Medical Center New York, NY, USA
| | - Yusuke Hirabayashi
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Kavli Institute for Brain Science, Columbia University Medical Center New York, NY, USA
| | - Franck Polleux
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Kavli Institute for Brain Science, Columbia University Medical Center New York, NY, USA
| |
Collapse
|
82
|
Deregulation of mitochondrial F1FO-ATP synthase via OSCP in Alzheimer's disease. Nat Commun 2016; 7:11483. [PMID: 27151236 PMCID: PMC5494197 DOI: 10.1038/ncomms11483] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 03/31/2016] [Indexed: 01/10/2023] Open
Abstract
F1FO-ATP synthase is critical for mitochondrial functions. The deregulation of this enzyme results in dampened mitochondrial oxidative phosphorylation (OXPHOS) and activated mitochondrial permeability transition (mPT), defects which accompany Alzheimer’s disease (AD). However, the molecular mechanisms that connect F1FO-ATP synthase dysfunction and AD remain unclear. Here, we observe selective loss of the oligomycin sensitivity conferring protein (OSCP) subunit of the F1FO-ATP synthase and the physical interaction of OSCP with amyloid beta (Aβ) in the brains of AD individuals and in an AD mouse model. Changes in OSCP levels are more pronounced in neuronal mitochondria. OSCP loss and its interplay with Aβ disrupt F1FO-ATP synthase, leading to reduced ATP production, elevated oxidative stress and activated mPT. The restoration of OSCP ameliorates Aβ-mediated mouse and human neuronal mitochondrial impairments and the resultant synaptic injury. Therefore, mitochondrial F1FO-ATP synthase dysfunction associated with AD progression could potentially be prevented by OSCP stabilization. F1FO ATP synthase is a critical enzyme for the maintenance of mitochondrial function. Here the authors demonstrate that loss of the F1FO-ATP synthase subunit OSCP and the interaction of OSCP with Aβ peptide in Alzheimer’s disease patients and mouse models lead to F1FO-ATP synthase deregulation and disruption of synaptic mitochondrial function.
Collapse
|
83
|
Wang L, Guo L, Lu L, Sun H, Shao M, Beck SJ, Li L, Ramachandran J, Du Y, Du H. Synaptosomal Mitochondrial Dysfunction in 5xFAD Mouse Model of Alzheimer's Disease. PLoS One 2016; 11:e0150441. [PMID: 26942905 PMCID: PMC4778903 DOI: 10.1371/journal.pone.0150441] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/13/2016] [Indexed: 11/23/2022] Open
Abstract
Brain mitochondrial dysfunction is hallmark pathology of Alzheimer’s disease (AD). Recently, the role of synaptosomal mitochondrial dysfunction in the development of synaptic injury in AD has received increasing attention. Synaptosomal mitochondria are a subgroup of neuronal mitochondria specifically locating at synapses. They play an essential role in fueling synaptic functions by providing energy on the site; and their defects may lead to synaptic failure, which is an early and pronounced pathology in AD. In our previous studies we have determined early synaptosomal mitochondrial dysfunction in an AD animal model (J20 line) overexpressing human Amyloid beta (Aβ), the key mediator of AD. In view of the limitations of J20 line mice in representing the full aspects of amyloidopathy in AD cases, we employed 5xFAD mice which are thought to be a desirable paradigm of amyloidopathy as seen in AD subjects. In addition, we have also examined the status of synaptosomal mitochondrial dynamics as well as Parkin-mediated mitophagy which have not been previously investigated in this mouse model. In comparison to nontransgenic (nonTg mice), 5xFAD mice demonstrated prominent synaptosomal mitochondrial dysfunction. Moreover, synaptosomal mitochondria from the AD mouse model displayed imbalanced mitochondrial dynamics towards fission along with activated Parkin and LC3BII recruitment correlating to spatial learning & memory impairments in 5xFAD mice in an age-dependent manner. These results suggest that synaptosomal mitochondrial deficits are primary pathology in Aβ-rich environments and further confirm the relevance of synaptosomal mitochondrial deficits to the development of AD.
Collapse
Affiliation(s)
- Lu Wang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
- Shandong University, Shandong Provincial Hospital, Jinan, Shandong Province, China, 250100
| | - Lan Guo
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
| | - Lin Lu
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
- Shandong University, Shandong Provincial Hospital, Jinan, Shandong Province, China, 250100
| | - Huili Sun
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
- Shenzhen Traditional Medicine Hospital, Shenzhen, Guangdong Province, China, 518031
| | - Muming Shao
- Shenzhen Traditional Medicine Hospital, Shenzhen, Guangdong Province, China, 518031
| | - Simon J. Beck
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
| | - Lin Li
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
| | - Janani Ramachandran
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
| | - Yifeng Du
- Shandong University, Shandong Provincial Hospital, Jinan, Shandong Province, China, 250100
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, United States of America, 75080
- Shandong University, Shandong Provincial Hospital, Jinan, Shandong Province, China, 250100
- * E-mail:
| |
Collapse
|
84
|
Lores-Arnaiz S, Lombardi P, Karadayian AG, Orgambide F, Cicerchia D, Bustamante J. Brain cortex mitochondrial bioenergetics in synaptosomes and non-synaptic mitochondria during aging. Neurochem Res 2016; 41:353-63. [DOI: 10.1007/s11064-015-1817-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/23/2015] [Accepted: 12/25/2015] [Indexed: 10/22/2022]
|
85
|
Scheff SW, Price DA, Ansari MA, Roberts KN, Schmitt FA, Ikonomovic MD, Mufson EJ. Synaptic change in the posterior cingulate gyrus in the progression of Alzheimer's disease. J Alzheimers Dis 2015; 43:1073-90. [PMID: 25147118 DOI: 10.3233/jad-141518] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mild cognitive impairment (MCI) is considered to be an early stage in the progression of Alzheimer's disease (AD) providing an opportunity to investigate brain pathogenesis prior to the onset of dementia. Neuroimaging studies have identified the posterior cingulate gyrus (PostC) as a cortical region affected early in the onset of AD. This association cortex is involved in a variety of different cognitive tasks and is intimately connected with the hippocampal/entorhinal cortex region, a component of the medial temporal memory circuit that displays early AD pathology. We quantified the total number of synapses in lamina 3 of the PostC using unbiased stereology coupled with electron microscopy from short postmortem autopsy tissue harvested from cases at different stage of AD progression. Individuals in the early stages of AD showed a significant decline in synaptic numbers compared to individuals with no cognitive impairment (NCI). Subjects with MCI exhibited synaptic numbers that were between the AD and NCI cohorts. Adjacent tissue was evaluated for changes in both pre and postsynaptic proteins levels. Individuals with MCI demonstrated a significant loss in presynaptic markers synapsin-1 and synaptophysin and postsynaptic markers PSD-95 and SAP-97. Levels of [3H]PiB binding was significantly increased in MCI and AD and correlated strongly with levels of synaptic proteins. All synaptic markers showed a significant association with Mini-Mental Status Examination scores. These results support the idea that the PostC synaptic function is affected during the prodromal stage of the disease and may underlie some of the early clinical sequelae associated with AD.
Collapse
Affiliation(s)
- Stephen W Scheff
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Douglas A Price
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Mubeen A Ansari
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Kelly N Roberts
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Elliott J Mufson
- Rush University Medical Center, Department of Neurological Sciences, Chicago, IL, USA
| |
Collapse
|
86
|
Bujdoso R, Landgraf M, Jackson WS, Thackray AM. Prion-induced neurotoxicity: Possible role for cell cycle activity and DNA damage response. World J Virol 2015; 4:188-197. [PMID: 26279981 PMCID: PMC4534811 DOI: 10.5501/wjv.v4.i3.188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 03/19/2015] [Accepted: 04/30/2015] [Indexed: 02/05/2023] Open
Abstract
Protein misfolding neurodegenerative diseases arise through neurotoxicity induced by aggregation of host proteins. These conditions include Alzheimer’s disease, Huntington’s disease, Parkinson’s disease, motor neuron disease, tauopathies and prion diseases. Collectively, these conditions are a challenge to society because of the increasing aged population and through the real threat to human food security by animal prion diseases. It is therefore important to understand the cellular and molecular mechanisms that underlie protein misfolding-induced neurotoxicity as this will form the basis for designing strategies to alleviate their burden. Prion diseases are an important paradigm for neurodegenerative conditions in general since several of these maladies have now been shown to display prion-like phenomena. Increasingly, cell cycle activity and the DNA damage response are recognised as cellular events that participate in the neurotoxic process of various neurodegenerative diseases, and their associated animal models, which suggests they are truly involved in the pathogenic process and are not merely epiphenomena. Here we review the role of cell cycle activity and the DNA damage response in neurodegeneration associated with protein misfolding diseases, and suggest that these events contribute towards prion-induced neurotoxicity. In doing so, we highlight PrP transgenic Drosophila as a tractable model for the genetic analysis of transmissible mammalian prion disease.
Collapse
|
87
|
Abstract
During synaptic vesicle (SV) recycling, the vacuolar-type H(+) ATPase creates a proton electrochemical gradient (ΔμH(+)) that drives neurotransmitter loading into SVs. Given the low estimates of free luminal protons, it has been envisioned that the influx of a limited number of protons suffices to establish ΔμH(+). Consistent with this, the time constant of SV re-acidification was reported to be <5 s, much faster than glutamate loading (τ of ∼ 15 s) and thus unlikely to be rate limiting for neurotransmitter loading. However, such estimates have relied on pHluorin-based probes that lack sensitivity in the lower luminal pH range. Here, we reexamined re-acidification kinetics using the mOrange2-based probe that should report the SV pH more accurately. In recordings from cultured mouse hippocampal neurons, we found that re-acidification took substantially longer (τ of ∼ 15 s) than estimated previously. In addition, we found that the SV lumen exhibited a large buffering capacity (∼ 57 mm/pH), corresponding to an accumulation of ∼ 1200 protons during re-acidification. Together, our results uncover hitherto unrecognized robust proton influx and storage in SVs that can restrict the rate of neurotransmitter refilling.
Collapse
|
88
|
Zhang XL, Guariglia SR, McGlothan JL, Stansfield KH, Stanton PK, Guilarte TR. Presynaptic mechanisms of lead neurotoxicity: effects on vesicular release, vesicle clustering and mitochondria number. PLoS One 2015; 10:e0127461. [PMID: 26011056 PMCID: PMC4444102 DOI: 10.1371/journal.pone.0127461] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/15/2015] [Indexed: 01/12/2023] Open
Abstract
Childhood lead (Pb2+) intoxication is a global public health problem and accounts for 0.6% of the global burden of disease associated with intellectual disabilities. Despite the recognition that childhood Pb2+ intoxication contributes significantly to intellectual disabilities, there is a fundamental lack of knowledge on presynaptic mechanisms by which Pb2+ disrupts synaptic function. In this study, using a well-characterized rodent model of developmental Pb2+ neurotoxicity, we show that Pb2+ exposure markedly inhibits presynaptic vesicular release in hippocampal Schaffer collateral-CA1 synapses in young adult rats. This effect was associated with ultrastructural changes which revealed a reduction in vesicle number in the readily releasable/docked vesicle pool, disperse vesicle clusters in the resting pool, and a reduced number of presynaptic terminals with multiple mitochondria with no change in presynaptic calcium influx. These studies provide fundamental knowledge on mechanisms by which Pb2+ produces profound inhibition of presynaptic vesicular release that contribute to deficits in synaptic plasticity and intellectual development.
Collapse
Affiliation(s)
- Xiao-lei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Sara R. Guariglia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Jennifer L. McGlothan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Kirstie H. Stansfield
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Patric K. Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Tomás R. Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
89
|
Behaviour and prefrontal protein differences in C57BL/6N and 129 X1/SvJ mice. Brain Res Bull 2015; 116:16-24. [PMID: 26003851 DOI: 10.1016/j.brainresbull.2015.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/24/2023]
Abstract
Experimental animals provide valuable opportunities to establish aetiological mechanisms and test new treatments for neurodevelopmental psychiatric conditions. However, it is increasingly appreciated that inter-strain differences cannot be neglected in the experimental design. In addition, the importance of including females in preclinical - but also clinical - research is now recognised. Here, we compared behaviour and prefrontal protein differences in male and female C57BL/6N and 129X1/SvJ mice as both are commonly used experimental rodents. Relative to 129X1/SvJ mice, both sexes of C57BL/6N mice had weaker sensorimotor gating, measured in the prepulse inhibition (PPI) of startle paradigm, and were more sensitive to amphetamine challenge in the open field. The pattern of protein expression in the prefrontal cortex of C57BL6N mice was also clearly distinct from 129X1/SvJ mice. Proteins differentially expressed were those associated with oxidative metabolism, receptor protein signalling, cell communication and signal transduction and energy pathways. We suggest that the C57BL/6N mouse may usefully proxy features of the neurodevelopmental disorders and could have application in pre-translational screening of new therapeutic approaches. The 129X1/SvJ strain in contrast, might be better suited to experimental studies of causal risk factors expected to lower PPI and increase amphetamine sensitivity.
Collapse
|
90
|
Guo Y, Menezes MJ, Menezes MP, Liang J, Li D, Riley LG, Clarke NF, Andrews PI, Tian L, Webster R, Wang F, Liu X, Shen Y, Thorburn DR, Keating BJ, Engel A, Hakonarson H, Christodoulou J, Xu X. Delayed diagnosis of congenital myasthenia due to associated mitochondrial enzyme defect. Neuromuscul Disord 2014; 25:257-61. [PMID: 25557462 DOI: 10.1016/j.nmd.2014.11.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/15/2014] [Accepted: 11/24/2014] [Indexed: 11/18/2022]
Abstract
Clinical phenotypes of congenital myasthenic syndromes and primary mitochondrial disorders share significant overlap in their clinical presentations, leading to challenges in making the correct diagnosis. Next generation sequencing is transforming molecular diagnosis of inherited neuromuscular disorders by identifying novel disease genes and by identifying previously known genes in undiagnosed patients. This is evident in two patients who were initially suspected to have a mitochondrial myopathy, but in whom a clear diagnosis of congenital myasthenic syndromes was made through whole exome sequencing. In patient 1, whole exome sequencing revealed compound heterozygous mutations c.1228C > T (p.Arg410Trp) and c.679C > T (p.Arg227*) in collagen-like tail subunit (single strand of homotrimer) of asymmetric acetylcholinesterase (COLQ). In patient 2, in whom a deletion of exon 52 in Dystrophin gene was previously detected by multiplex ligation-dependent probe amplification, Sanger sequencing revealed an additional homozygous mutation c.1511_1513delCTT (p.Pro504Argfs*183) in docking protein7 (DOK7). These case reports highlight the need for careful diagnosis of clinically heterogeneous syndromes like congenital myasthenic syndromes, which are treatable, and for which delayed diagnosis is likely to have implications for patient health. The report also demonstrates that whole exome sequencing is an effective diagnostic tool in providing molecular diagnosis in patients with complex phenotypes.
Collapse
Affiliation(s)
- Yiran Guo
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Minal J Menezes
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics & Child Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Manoj P Menezes
- Discipline of Paediatrics & Child Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | | | - Dong Li
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Lisa G Riley
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Nigel F Clarke
- Discipline of Paediatrics & Child Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - P Ian Andrews
- Department of Neurology, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Lifeng Tian
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Richard Webster
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Fengxiang Wang
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | | | | | - David R Thorburn
- Murdoch Childrens Research Institute and Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Brendan J Keating
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Human Genetics Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Andrew Engel
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Human Genetics Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - John Christodoulou
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, NSW, Australia; Discipline of Paediatrics & Child Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Discipline of Genetic Medicine, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China; The Guangdong Enterprise Key Laboratory of Human Disease Genomics, BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
91
|
Celardo I, Martins LM, Gandhi S. Unravelling mitochondrial pathways to Parkinson's disease. Br J Pharmacol 2014; 171:1943-57. [PMID: 24117181 PMCID: PMC3976614 DOI: 10.1111/bph.12433] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/09/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are essential for cellular function due to their role in ATP production, calcium homeostasis and apoptotic signalling. Neurons are heavily reliant on mitochondrial integrity for their complex signalling, plasticity and excitability properties, and to ensure cell survival over decades. The maintenance of a pool of healthy mitochondria that can meet the bioenergetic demands of a neuron, is therefore of critical importance; this is achieved by maintaining a careful balance between mitochondrial biogenesis, mitochondrial trafficking, mitochondrial dynamics and mitophagy. The molecular mechanisms that underlie these processes are gradually being elucidated. It is widely recognized that mitochondrial dysfunction occurs in many neurodegenerative diseases, including Parkinson's disease. Mitochondrial dysfunction in the form of reduced bioenergetic capacity, increased oxidative stress and reduced resistance to stress, is observed in several Parkinson's disease models. However, identification of the recessive genes implicated in Parkinson's disease has revealed a common pathway involving mitochondrial dynamics, transport, turnover and mitophagy. This body of work has led to the hypothesis that the homeostatic mechanisms that ensure a healthy mitochondrial pool are key to neuronal function and integrity. In this paradigm, impaired mitochondrial dynamics and clearance result in the accumulation of damaged and dysfunctional mitochondria, which may directly induce neuronal dysfunction and death. In this review, we consider the mechanisms by which mitochondrial dysfunction may lead to neurodegeneration. In particular, we focus on the mechanisms that underlie mitochondrial homeostasis, and discuss their importance in neuronal integrity and neurodegeneration in Parkinson's disease.
Collapse
|
92
|
Rappold PM, Cui M, Grima JC, Fan RZ, de Mesy-Bentley KL, Chen L, Zhuang X, Bowers WJ, Tieu K. Drp1 inhibition attenuates neurotoxicity and dopamine release deficits in vivo. Nat Commun 2014; 5:5244. [PMID: 25370169 PMCID: PMC4223875 DOI: 10.1038/ncomms6244] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/10/2014] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction has been reported in both familial and sporadic Parkinson's disease (PD). However, effective therapy targeting this pathway is currently inadequate. Recent studies suggest that manipulating the processes of mitochondrial fission and fusion has considerable potential for treating human diseases. To determine the therapeutic impact of targeting these pathways on PD, we used two complementary mouse models of mitochondrial impairments as seen in PD. We show here that blocking mitochondrial fission is neuroprotective in the PTEN-induced putative kinase-1 deletion (PINK1(-/-)) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse models. Specifically, we show that inhibition of the mitochondrial fission GTPase dynamin-related protein-1 (Drp1) using gene-based and small-molecule approaches attenuates neurotoxicity and restores pre-existing striatal dopamine release deficits in these animal models. These results suggest Drp1 inhibition as a potential treatment for PD.
Collapse
Affiliation(s)
- Phillip M Rappold
- Department of Environmental Medicine, Center for Translational Neuromedicine, University of Rochester School of Medicine, 575 Elmwood Avenue, Rochester, New York 14642, USA
| | - Mei Cui
- Department of Environmental Medicine, Center for Translational Neuromedicine, University of Rochester School of Medicine, 575 Elmwood Avenue, Rochester, New York 14642, USA
| | - Jonathan C Grima
- Department of Environmental Medicine, Center for Translational Neuromedicine, University of Rochester School of Medicine, 575 Elmwood Avenue, Rochester, New York 14642, USA
| | - Rebecca Z Fan
- Department of Clinical Neurobiology and Institute of Translational and Stratified Medicine, Plymouth University, John Bull Building, Plymouth PL6 8BU, UK
| | - Karen L de Mesy-Bentley
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York 14642, USA
| | - Linan Chen
- Department of Neurobiology, University of Chicago, 947 E. 58th Street, Chicago, Illinois 60637, USA
| | - Xiaoxi Zhuang
- Department of Neurobiology, University of Chicago, 947 E. 58th Street, Chicago, Illinois 60637, USA
| | - William J Bowers
- Department of Neurology, Center for Neural Development and Disease, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, New York 14642, USA
| | - Kim Tieu
- 1] Department of Environmental Medicine, Center for Translational Neuromedicine, University of Rochester School of Medicine, 575 Elmwood Avenue, Rochester, New York 14642, USA [2] Department of Clinical Neurobiology and Institute of Translational and Stratified Medicine, Plymouth University, John Bull Building, Plymouth PL6 8BU, UK
| |
Collapse
|
93
|
Mendoza E, Miranda-Barrientos J, Vázquez-Roque R, Morales-Herrera E, Ruelas A, De la Rosa G, Flores G, Hernández-Echeagaray E. In vivo mitochondrial inhibition alters corticostriatal synaptic function and the modulatory effects of neurotrophins. Neuroscience 2014; 280:156-70. [DOI: 10.1016/j.neuroscience.2014.09.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 09/09/2014] [Indexed: 01/09/2023]
|
94
|
Mitochondrial dysfunction in schizophrenia: an evolutionary perspective. Hum Genet 2014; 134:13-21. [PMID: 25312050 DOI: 10.1007/s00439-014-1491-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 09/26/2014] [Indexed: 10/24/2022]
Abstract
Schizophrenia (SCZ) is a severe psychiatric illness with a lifetime prevalence of 0.4 %. A disturbance of energy metabolism has been suggested as part of the etiopathogenesis of the disorder. Several lines of evidence have proposed a connection between etiopathogenesis of SCZ and human brain evolution, which was characterized by an increase in the energy requirement, demanding a co-evolution of the mitochondrial system. Mitochondria are key players in brain energy homeostasis and multiple lines of evidence suggest that the system is disrupted in SCZ. In this review, we will describe the current knowledge on pathways/system involved in the human brain evolution as well as the main theories regarding the evolutionary origin of SCZ. We will furthermore discuss the role of mitochondria in the context of brain energy metabolism and its role in the etiopathogenesis of SCZ. Understanding SCZ in the context of human brain evolution opens a new perspective to elucidate pathophysiological mechanisms involved in the origin and/or portions of the complex symptomatology of this severe mental disorder.
Collapse
|
95
|
Dukoff DJ, Hogg DW, Hawrysh PJ, Buck LT. Scavenging ROS dramatically increase NMDA receptor whole-cell currents in painted turtle cortical neurons. ACTA ACUST UNITED AC 2014; 217:3346-55. [PMID: 25063855 DOI: 10.1242/jeb.105825] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Oxygen deprivation triggers excitotoxic cell death in mammal neurons through excessive calcium loading via over-activation of N-methyl-d-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. This does not occur in the western painted turtle, which overwinters for months without oxygen. Neurological damage is avoided through anoxia-mediated decreases in NMDA and AMPA receptor currents that are dependent upon a modest rise in intracellular Ca(2+) concentrations ([Ca(2+)]i) originating from mitochondria. Anoxia also blocks mitochondrial reactive oxygen species (ROS) generation, which is another potential signaling mechanism to regulate glutamate receptors. To assess the effects of decreased intracellular [ROS] on NMDA and AMPA receptor currents, we scavenged ROS with N-2-mercaptopropionylglycine (MPG) or N-acetylcysteine (NAC). Unlike anoxia, ROS scavengers increased NMDA receptor whole-cell currents by 100%, while hydrogen peroxide decreased currents. AMPA receptor currents and [Ca(2+)]i concentrations were unaffected by ROS manipulation. Because decreases in [ROS] increased NMDA receptor currents, we next asked whether mitochondrial Ca(2+) release prevents receptor potentiation during anoxia. Normoxic activation of mitochondrial ATP-sensitive potassium (mKATP) channels with diazoxide decreased NMDA receptor currents and was unaffected by subsequent ROS scavenging. Diazoxide application following ROS scavenging did not rescue scavenger-mediated increases in NMDA receptor currents. Fluorescent measurement of [Ca(2+)]i and ROS levels demonstrated that [Ca(2+)]i increases before ROS decreases. We conclude that decreases in ROS concentration are not linked to anoxia-mediated decreases in NMDA/AMPA receptor currents but are rather associated with an increase in NMDA receptor currents that is prevented during anoxia by mitochondrial Ca(2+) release.
Collapse
Affiliation(s)
- David James Dukoff
- Department of Cell and Systems Biology and Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - David William Hogg
- Department of Cell and Systems Biology and Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Peter John Hawrysh
- Department of Cell and Systems Biology and Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Leslie Thomas Buck
- Department of Cell and Systems Biology and Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
96
|
Shi Y, Ivannikov MV, Walsh ME, Liu Y, Zhang Y, Jaramillo CA, Macleod GT, Van Remmen H. The lack of CuZnSOD leads to impaired neurotransmitter release, neuromuscular junction destabilization and reduced muscle strength in mice. PLoS One 2014; 9:e100834. [PMID: 24971750 PMCID: PMC4074103 DOI: 10.1371/journal.pone.0100834] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/29/2014] [Indexed: 11/29/2022] Open
Abstract
Elevated reactive oxygen species (ROS) production and ROS-dependent protein damage is a common observation in the pathogenesis of many muscle wasting disorders, including sarcopenia. However, the contribution of elevated ROS levels to –a breakdown in neuromuscular communication and muscle atrophy remains unknown. In this study, we examined a copper zinc superoxide dismutase [CuZnSOD (Sod1)] knockout mouse (Sod1−/−), a mouse model of elevated oxidative stress that exhibits accelerated loss of muscle mass, which recapitulates many phenotypes of sarcopenia as early as 5 months of age. We found that young adult Sod1−/− mice display a considerable reduction in hind limb skeletal muscle mass and strength when compared to age-matched wild-type mice. These changes are accompanied by gross alterations in neuromuscular junction (NMJ) morphology, including reduced occupancy of the motor endplates by axons, terminal sprouting and axon thinning and irregular swelling. Surprisingly however, the average density of acetylcholine receptors in endplates is preserved. Using in vivo electromyography and ex vivo electrophysiological studies of hind limb muscles in Sod1−/− mice, we found that motor axons innervating the extensor digitorum longus (EDL) and gastrocnemius muscles release fewer synaptic vesicles upon nerve stimulation. Recordings from individually identified EDL NMJs show that reductions in neurotransmitter release are apparent in the Sod1−/− mice even when endplates are close to fully innervated. However, electrophysiological properties, such as input resistance, resting membrane potential and spontaneous neurotransmitter release kinetics (but not frequency) are similar between EDL muscles of Sod1−/− and wild-type mice. Administration of the potassium channel blocker 3,4-diaminopyridine, which broadens the presynaptic action potential, improves both neurotransmitter release and muscle strength. Together, these results suggest that ROS-associated motor nerve terminal dysfunction is a contributor to the observed muscle changes in Sod1−/− mice.
Collapse
Affiliation(s)
- Yun Shi
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Maxim V. Ivannikov
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Michael E. Walsh
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yuhong Liu
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Yiqiang Zhang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Carlos A. Jaramillo
- Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Rehabilitation Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Gregory T. Macleod
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Holly Van Remmen
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
97
|
Laßek M, Weingarten J, Volknandt W. The Proteome of the Murine Presynaptic Active Zone. Proteomes 2014; 2:243-257. [PMID: 28250380 PMCID: PMC5302740 DOI: 10.3390/proteomes2020243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/09/2014] [Accepted: 04/21/2014] [Indexed: 01/09/2023] Open
Abstract
The proteome of the presynaptic active zone controls neurotransmitter release and the short- and long-term structural and functional dynamics of the nerve terminal. The proteinaceous inventory of the presynaptic active zone has recently been reported. This review will evaluate the subcellular fractionation protocols and the proteomic approaches employed. A breakthrough for the identification of the proteome of the presynaptic active zone was the successful employment of antibodies directed against a cytosolic epitope of membrane integral synaptic vesicle proteins for the immunopurification of synaptic vesicles docked to the presynaptic plasma membrane. Combining immunopurification and subsequent analytical mass spectrometry, hundreds of proteins, including synaptic vesicle proteins, components of the presynaptic fusion and retrieval machinery, proteins involved in intracellular and extracellular signaling and a large variety of adhesion molecules, were identified. Numerous proteins regulating the rearrangement of the cytoskeleton are indicative of the functional and structural dynamics of the presynapse. This review will critically discuss both the experimental approaches and prominent protein candidates identified. Many proteins have not previously been assigned to the presynaptic release sites and may be directly involved in the short- and long-term structural modulation of the presynaptic compartment. The identification of proteinaceous constituents of the presynaptic active zone provides the basis for further analyzing the interaction of presynaptic proteins with their targets and opens novel insights into the functional role of these proteins in neuronal communication.
Collapse
Affiliation(s)
- Melanie Laßek
- Institute for Cell Biology and Neuroscience, Department Molecular and Cellular Neurobiology, Max von Laue Str. 13, 60438 Frankfurt am Main, Germany.
| | - Jens Weingarten
- Institute for Cell Biology and Neuroscience, Department Molecular and Cellular Neurobiology, Max von Laue Str. 13, 60438 Frankfurt am Main, Germany.
| | - Walter Volknandt
- Institute for Cell Biology and Neuroscience, Department Molecular and Cellular Neurobiology, Max von Laue Str. 13, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
98
|
Rangaraju V, Calloway N, Ryan TA. Activity-driven local ATP synthesis is required for synaptic function. Cell 2014; 156:825-35. [PMID: 24529383 DOI: 10.1016/j.cell.2013.12.042] [Citation(s) in RCA: 551] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 11/01/2013] [Accepted: 12/31/2013] [Indexed: 01/20/2023]
Abstract
Cognitive function is tightly related to metabolic state, but the locus of this control is not well understood. Synapses are thought to present large ATP demands; however, it is unclear how fuel availability and electrical activity impact synaptic ATP levels and how ATP availability controls synaptic function. We developed a quantitative genetically encoded optical reporter of presynaptic ATP, Syn-ATP, and find that electrical activity imposes large metabolic demands that are met via activity-driven control of both glycolysis and mitochondrial function. We discovered that the primary source of activity-driven metabolic demand is the synaptic vesicle cycle. In metabolically intact synapses, activity-driven ATP synthesis is well matched to the energetic needs of synaptic function, which, at steady state, results in ∼10(6) free ATPs per nerve terminal. Despite this large reservoir of ATP, we find that several key aspects of presynaptic function are severely impaired following even brief interruptions in activity-stimulated ATP synthesis.
Collapse
Affiliation(s)
- Vidhya Rangaraju
- Rockefeller/Sloan-Kettering/Weill Cornell Tri-Institutional Training Program in Chemical Biology, New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Nathaniel Calloway
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
99
|
Pereira JC, Andersen ML. The role of thyroid hormone in sleep deprivation. Med Hypotheses 2014; 82:350-5. [DOI: 10.1016/j.mehy.2014.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/20/2013] [Accepted: 01/06/2014] [Indexed: 11/24/2022]
|
100
|
Weingarten J, Lassek M, Mueller BF, Rohmer M, Lunger I, Baeumlisberger D, Dudek S, Gogesch P, Karas M, Volknandt W. The proteome of the presynaptic active zone from mouse brain. Mol Cell Neurosci 2014; 59:106-18. [PMID: 24534009 DOI: 10.1016/j.mcn.2014.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/05/2014] [Accepted: 02/07/2014] [Indexed: 01/07/2023] Open
Abstract
Neurotransmitter release as well as the structural and functional dynamics of the presynaptic active zone is controlled by proteinaceous components. Here we describe for the first time an experimental approach for the isolation of the presynaptic active zone from individual mouse brains, a prerequisite for understanding the functional inventory of the presynaptic protein network and for the later analysis of changes occurring in mutant mice. Using a monoclonal antibody against the ubiquitous synaptic vesicle protein SV2 we immunopurified synaptic vesicles docked to the presynaptic plasma membrane. Enrichment studies by means of Western blot analysis and mass spectrometry identified 485 proteins belonging to an impressive variety of functional categories. Our data suggest that presynaptic active zones represent focal hot spots that are not only involved in the regulation of neurotransmitter release but also in multiple structural and functional alterations the adult nerve terminal undergoes during neural activity in adult CNS. They furthermore open new avenues for characterizing alterations in the active zone proteome of mutant mice and their corresponding controls, including the various mouse models of neurological diseases.
Collapse
Affiliation(s)
- Jens Weingarten
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Melanie Lassek
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Benjamin F Mueller
- Institute of Pharmaceutical Chemistry, Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| | - Marion Rohmer
- Institute of Pharmaceutical Chemistry, Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| | - Ilaria Lunger
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | | | - Simone Dudek
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Patricia Gogesch
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany
| | - Michael Karas
- Institute of Pharmaceutical Chemistry, Cluster of Excellence "Macromolecular Complexes", Goethe-University, Frankfurt am Main, Germany
| | - Walter Volknandt
- Institute for Cell Biology and Neuroscience, Biologicum, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|