1
|
Nordhjem BJT, Hjalgrim LL. Cancer-related cognitive impairment and hippocampal functioning: The role of dynamin-1. Neurotherapeutics 2025; 22:e00508. [PMID: 39676022 PMCID: PMC11742813 DOI: 10.1016/j.neurot.2024.e00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Affiliation(s)
| | - Lisa Lyngsie Hjalgrim
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
2
|
Ng DQ, Hudson C, Nguyen T, Gupta SK, Koh YQ, Acharya MM, Chan A. Dynamin-1 is a potential mediator in cancer-related cognitive impairment. Neurotherapeutics 2025; 22:e00480. [PMID: 39516074 PMCID: PMC11742811 DOI: 10.1016/j.neurot.2024.e00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Dynamin-1 (DNM1) is crucial for synaptic activity, neurotransmission, and associative memory, positioning it as a potential biomarker of cancer-related cognitive impairment (CRCI), a neurological consequence of cancer treatment characterized by memory loss, poor concentration, and impaired executive function. Through a stepwise approach, this study investigated the role of DNM1 in CRCI pathogenesis, incorporating both human data and animal models. The human study recruited newly diagnosed, chemotherapy-naïve adolescent and young adult cancer and non-cancer controls to complete a cognitive instrument (FACT-Cog) and blood draws for up to three time points. Following that, a syngeneic young-adult WT (C57BL/6) female mouse model of breast cancer chemobrain was developed to study DNM1 expression in the hippocampus. Samples from eighty-six participants with 30 adolescent and young adult (AYA) cancer and 56 non-cancer participants were analyzed. DNM1 levels were 32 % lower (P = 0.041) among cancer participants compared to non-cancer prior to treatment. After receiving cytotoxic treatment, cognitively impaired cancer patients were found to have 46 % lower DNM1 levels than those without impairment (P = 0.049). In murine breast cancer-bearing mice receiving chemotherapy, we found a greater than 40 % decline (P < 0.0001) in DNM1 immunoreactivity in the hippocampal CA1 and CA3 subregions concurrent with a deterioration in spatial recognition memory (P < 0.02), compared to control mice without exposure to cancer and chemotherapy. Consistently observed in both human and animal studies, the downregulation of DNM1 is linked with the onset of CRCI. DNM1 might be a biomarker and therapeutic target for CRCI.
Collapse
Affiliation(s)
- Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Casey Hudson
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Tracy Nguyen
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Sukesh Kumar Gupta
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Yong Qin Koh
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, USA; Department of Radiation Oncology, School of Medicine, University of California Irvine, Irvine, CA, USA.
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA; Department of Oncology Pharmacy, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
3
|
Ng DQ, Hudson C, Nguyen T, Gupta SK, Koh YQ, Acharya MM, Chan A. Dynamin-1 is a potential mediator in Cancer-Related Cognitive Impairment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597349. [PMID: 38895481 PMCID: PMC11185648 DOI: 10.1101/2024.06.04.597349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Dynamin-1 (DNM1) consolidates memory through synaptic transmission and modulation and has been explored as a therapeutic target in Alzheimer's disease. Through a two-prong approach, this study examined its role in cancer-related cognitive impairment (CRCI) pathogenesis using human and animal models. The human study recruited newly diagnosed, chemotherapy-naïve adolescent and young adult cancer and non-cancer controls to complete a cognitive instrument (FACT-Cog) and blood draws for up to three time points. Concurrently, a syngeneic young-adult WT (C57BL/6 female) mouse model of breast cancer was developed to study DNM1 expression in the brain. Samples from eighty-six participants with 30 adolescent and young adult (AYA) cancer and 56 non-cancer participants were analyzed. DNM1 levels were significantly lower among cancer participants compared to non-cancer prior to treatment. While receiving cancer treatment, cognitively impaired patients were found with a significant downregulation of DNM1, but not among those without impairment. In murine breast cancer-bearing mice receiving chemotherapy, we consistently found a significant decline in DNM1 immunoreactivity in the hippocampal CA1 and CA3 subregions. Observed in both human and animal studies, the downregulation of DNM1 is linked with the onset of CRCI. Future research should explore the potential of DNM1 in CRCI pathogenesis and therapeutics development.
Collapse
|
4
|
D'Acunzo P, Argyrousi EK, Ungania JM, Kim Y, DeRosa S, Pawlik M, Goulbourne CN, Arancio O, Levy E. Mitovesicles secreted into the extracellular space of brains with mitochondrial dysfunction impair synaptic plasticity. Mol Neurodegener 2024; 19:34. [PMID: 38616258 PMCID: PMC11017499 DOI: 10.1186/s13024-024-00721-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Hypometabolism tied to mitochondrial dysfunction occurs in the aging brain and in neurodegenerative disorders, including in Alzheimer's disease, in Down syndrome, and in mouse models of these conditions. We have previously shown that mitovesicles, small extracellular vesicles (EVs) of mitochondrial origin, are altered in content and abundance in multiple brain conditions characterized by mitochondrial dysfunction. However, given their recent discovery, it is yet to be explored what mitovesicles regulate and modify, both under physiological conditions and in the diseased brain. In this study, we investigated the effects of mitovesicles on synaptic function, and the molecular players involved. METHODS Hippocampal slices from wild-type mice were perfused with the three known types of EVs, mitovesicles, microvesicles, or exosomes, isolated from the brain of a mouse model of Down syndrome or of a diploid control and long-term potentiation (LTP) recorded. The role of the monoamine oxidases type B (MAO-B) and type A (MAO-A) in mitovesicle-driven LTP impairments was addressed by treatment of mitovesicles with the irreversible MAO inhibitors pargyline and clorgiline prior to perfusion of the hippocampal slices. RESULTS Mitovesicles from the brain of the Down syndrome model reduced LTP within minutes of mitovesicle addition. Mitovesicles isolated from control brains did not trigger electrophysiological effects, nor did other types of brain EVs (microvesicles and exosomes) from any genotype tested. Depleting mitovesicles of their MAO-B, but not MAO-A, activity eliminated their ability to alter LTP. CONCLUSIONS Mitovesicle impairment of LTP is a previously undescribed paracrine-like mechanism by which EVs modulate synaptic activity, demonstrating that mitovesicles are active participants in the propagation of cellular and functional homeostatic changes in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Pasquale D'Acunzo
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 10016, New York, NY, USA
| | - Elentina K Argyrousi
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 10027, New York, NY, USA
- Department of Medicine, Columbia University, 10027, New York, NY, USA
| | - Jonathan M Ungania
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 10016, New York, NY, USA
| | - Steven DeRosa
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Monika Pawlik
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 10027, New York, NY, USA
- Department of Medicine, Columbia University, 10027, New York, NY, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, 10016, New York, NY, USA.
- Department of Biochemistry & Molecular Pharmacology, New York University Grossman School of Medicine, 10027, New York, NY, USA.
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 10016, New York, NY, USA.
| |
Collapse
|
5
|
Zhou S, Zhou Y, Zhong W, Su Z, Qin Z. Involvement of protein L-isoaspartyl methyltransferase in the physiopathology of neurodegenerative diseases: Possible substrates associated with synaptic function. Neurochem Int 2023; 170:105606. [PMID: 37657764 DOI: 10.1016/j.neuint.2023.105606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
Synaptic dysfunction is a typical pathophysiologic change in neurodegenerative diseases (NDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), Hintington's disease (HD) and amyotrophic lateral sclerosis (ALS), which involves protein post-translational modifications (PTMs) including L-isoaspartate (L-isoAsp) formed by isomerization of aspartate or deamidation of asparagine. The formation of L-isoAsp could be repaired by protein L-isoaspartyl methyltransferase (PIMT). Some synaptic proteins have been identified as PIMT potential substrates and play an essential role in ensuring synaptic function. In this review, we discuss the role of certain synaptic proteins as PIMT substrates in neurodegenerative disease, thus providing therapeutic synapse-centered targets for the treatment of NDs.
Collapse
Affiliation(s)
- Sirui Zhou
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yancheng Zhou
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wanyu Zhong
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhonghao Su
- Department of Febrile Disease, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhenxia Qin
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
6
|
Medina-Vera D, Enache D, Tambaro S, Abuhashish E, Rosell-Valle C, Winblad B, Rodríguez de Fonseca F, Bereczki E, Nilsson P. Translational potential of synaptic alterations in Alzheimer's disease patients and amyloid precursor protein knock-in mice. Brain Commun 2023; 5:fcad001. [PMID: 36687391 PMCID: PMC9851419 DOI: 10.1093/braincomms/fcad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 09/19/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Synaptic dysfunction is an early event in Alzheimer's disease. Post-mortem studies suggest that alterations in synaptic proteins are associated with cognitive decline in Alzheimer's disease. We measured the concentration of three synaptic proteins, zinc transporter protein 3, dynamin1 and AMPA glutamate receptor 3 in cerebrospinal fluid of subjects with mild cognitive impairment (n = 18) and Alzheimer's disease (n = 18) and compared the levels to cognitively and neurologically healthy controls (n = 18) by using ELISA assay. In addition, we aimed to assess the translational potential of these synaptic proteins in two established amyloid precursor protein knock-in Alzheimer's disease mouse models by assessing the cerebrospinal fluid, hippocampal and cortical synaptic protein concentrations. Using ELISA, we measured in parallel these three proteins in cerebrospinal fluid and/or brain of 12- and 24-month-old AppNL-F and AppNL-G-F knock-in mice and AppWt control mice. The regional distribution and expression of these proteins were explored upon aging of the App knock-in models by quantitative immunofluorescence microscopy. Notably, we found a significant increase in concentrations of zinc transporter protein 3 and AMPA glutamate receptor 3 in cerebrospinal fluid of both patient groups compared with cognitively healthy controls. Dynamin1 concentration was significantly higher in Alzheimer's disease patients. Remarkably, patients with mild cognitive impairment who converted to Alzheimer's disease (n = 7) within 2 years exhibited elevated baseline cerebrospinal fluid zinc transporter protein 3 concentrations compared with mild cognitive impairment patients who did not convert (n = 11). Interestingly, similar to the alterations in Alzheimer's disease subjects, cerebrospinal fluid AMPA glutamate receptor 3 concentration was significantly higher in AppNL-G-F knock-in mice when compared with wild-type controls. Furthermore, we have detected age and brain regional specific changes of the three synaptic proteins in the hippocampus and prefrontal cortex of both AppNL-F and AppNL-G-F knock-in mice. Notably, all the three cerebrospinal fluid synaptic protein concentrations correlated negatively with concentrations in hippocampal lysates. The elevated zinc transporter protein 3 concentrations in the cerebrospinal fluid of converter versus non-converter mild cognitive impairment patients suggests a prospective role of zinc transporter 3 in differentiating dementia patients of the biological continuum of Alzheimer's disease. The increased cerebrospinal fluid concentrations of synaptic proteins in both patient groups, potentially reflecting synaptic alterations in the brain, were similarly observed in the amyloid precursor protein knock-in mouse models highlighting the translational potential of these proteins as markers for synaptic alterations. These synaptic markers could potentially help reduce the current disparities between human and animal model-based studies aiding the translation of preclinical discoveries of pathophysiological changes into clinical research.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga 29010, Spain,Facultad de Ciencias, Universidad de Málaga, Málaga 29010, Spain,Facultad de Medicina, Universidad de Málaga, Málaga 29010, Spain
| | - Daniela Enache
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden
| | - Ethar Abuhashish
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden
| | - Cristina Rosell-Valle
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga 29010, Spain
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 17164 Solna, Sweden,Theme Inflammation and Aging, Karolinska University Hospital, 17164 Solna, Sweden
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Málaga 29010, Spain
| | - Erika Bereczki
- Correspondence to: Erika Bereczki Department of NVS, Center for Alzheimer Research Division of Neurogeriatrics, Karolinska Institutet BioClinicum J10:30, 17 164, Stockholm, Sweden E-mail:
| | | |
Collapse
|
7
|
Sadhu A, Badal KK, Zhao Y, Ali AA, Swarnkar S, Tsaprailis G, Crynen GC, Puthanveettil SV. Short-Term and Long-Term Sensitization Differentially Alters the Composition of an Anterograde Transport Complex in Aplysia. eNeuro 2023; 10:ENEURO.0266-22.2022. [PMID: 36549915 PMCID: PMC9829102 DOI: 10.1523/eneuro.0266-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/09/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Long-term memory formation requires anterograde transport of proteins from the soma of a neuron to its distal synaptic terminals. This allows new synaptic connections to be grown and existing ones remodeled. However, we do not yet know which proteins are transported to synapses in response to activity and temporal regulation. Here, using quantitative mass spectrometry, we have profiled anterograde protein cargos of a learning-regulated molecular motor protein kinesin [Aplysia kinesin heavy chain 1 (ApKHC1)] following short-term sensitization (STS) and long-term sensitization (LTS) in Aplysia californica Our results reveal enrichment of specific proteins associated with ApKHC1 following both STS and LTS, as well as temporal changes within 1 and 3 h of LTS training. A significant number of proteins enriched in the ApKHC1 complex participate in synaptic function, and, while some are ubiquitously enriched across training conditions, a few are enriched in response to specific training. For instance, factors aiding new synapse formation, such as synaptotagmin-1, dynamin-1, and calmodulin, are differentially enriched in anterograde complexes 1 h after LTS but are depleted 3 h after LTS. Proteins including gelsolin-like protein 2 and sec23A/sec24A, which function in actin filament stabilization and vesicle transport, respectively, are enriched in cargos 3 h after LTS. These results establish that the composition of anterograde transport complexes undergo experience-dependent specific changes and illuminate dynamic changes in the communication between soma and synapse during learning.
Collapse
Affiliation(s)
- Abhishek Sadhu
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Kerriann K Badal
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
- Integrated Biology Graduate Program, Florida Atlantic University, Jupiter, Florida 33458
| | - Yibo Zhao
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Adia A Ali
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Supriya Swarnkar
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - George Tsaprailis
- Proteomics Core, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Gogce C Crynen
- Bioinformatics Core, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | | |
Collapse
|
8
|
Sha S, Xing XN, Wang T, Li Y, Zhang RW, Shen XL, Cao YP, Qu L. DNA vaccines targeting amyloid-β oligomer ameliorate cognitive deficits of aged APP/PS1/tau triple-transgenic mouse models of Alzheimer's disease. Neural Regen Res 2022; 17:2305-2310. [PMID: 35259854 PMCID: PMC9083157 DOI: 10.4103/1673-5374.337054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The amyloid-β (Aβ) oligomer, rather than the Aβ monomer, is considered to be the primary initiator of Alzheimer’s disease. It was hypothesized that p(Aβ3–10)10-MT, the recombinant Aβ3–10 gene vaccine of the Aβ oligomer has the potential to treat Alzheimer’s disease. In this study, we intramuscularly injected the p(Aβ3–10)10-MT vaccine into the left hindlimb of APP/PS1/tau triple-transgenic mice, which are a model for Alzheimer’s disease. Our results showed that the p(Aβ3–10)10-MT vaccine effectively reduced Aβ oligomer levels and plaque deposition in the cerebral cortex and hippocampus, decreased the levels tau protein variants, reduced synaptic loss, protected synaptic function, reduced neuron loss, and ameliorated memory impairment without causing any cerebral hemorrhaging. Therefore, this novel DNA vaccine, which is safe and highly effective in mouse models of Alzheimer’s disease, holds a lot of promise for the treatment of Alzheimer’s disease in humans.
Collapse
Affiliation(s)
- Sha Sha
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiao-Na Xing
- Department of Neurology, Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Tao Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Ying Li
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Rong-Wei Zhang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xue-Li Shen
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yun-Peng Cao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Le Qu
- Department of Dermatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
9
|
Grochowalska K, Pikul P, Piwkowska A. Insights into the regulation of podocyte and glomerular function by lactate and its metabolic sensor G-protein-coupled receptor 81. J Cell Physiol 2022; 237:4097-4111. [PMID: 36084306 DOI: 10.1002/jcp.30874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 11/10/2022]
Abstract
Podocytes and their foot processes are an important cellular layer of the renal filtration barrier that is involved in regulating glomerular permeability. Disturbances of podocyte function play a central role in the development of proteinuria in diabetic nephropathy. The retraction and effacement of podocyte foot processes that form slit diaphragms are a common feature of proteinuria. Correlations between the retraction of foot processes and the development of proteinuria are not well understood. Unraveling peculiarities of podocyte energy metabolism notably under diabetic conditions will provide insights into the pathogenesis of diabetic nephropathy. Intracellular metabolism in the cortical area of podocytes is regulated by glycolysis, whereas energy balance in the central area is controlled by oxidative phosphorylation and glycolysis. High glucose concentrations were recently reported to force podocytes to switch from mitochondrial oxidative phosphorylation to glycolysis, resulting in lactic acidosis. In this review, we hypothesize that the lactate receptor G-protein-coupled receptor 81 (also known as hydroxycarboxylic acid receptor 81) may contribute to the control of podocyte function in both health and disease.
Collapse
Affiliation(s)
- Klaudia Grochowalska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Piotr Pikul
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland.,Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| |
Collapse
|
10
|
The Effects of Matcha and Decaffeinated Matcha on Learning, Memory and Proteomics of Hippocampus in Senescence-Accelerated (SAMP8) Mice. Nutrients 2022; 14:nu14061197. [PMID: 35334854 PMCID: PMC8952568 DOI: 10.3390/nu14061197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/03/2022] Open
Abstract
Although the benefits of the consumption of green tea and its components, including catechins and theanine, regarding aging, memory impairment and age-related cognitive decline have been investigated in senescence-accelerated prone mice (SAMP8), studies that simultaneously measured the kinds of proteins that vary in their expression due to the administration of green tea and its extracts were not found. In this study, the effect of dietary and decaffeinated matcha on protein expression in the hippocampus of SAMP 8 was examined comprehensively, mainly using proteomics. Although improvements in memory and the hair appearance of the back coat were limited upon administering the samples, the following regulations were observed in some of the proteins involved in neuron degeneration, Parkinson’s and Alzheimer’s diseases, synapse transmission and nerve cell plasticity, antioxidation, glutamate transport and metabolism, GABA (γ-amino butyric acid) formation and transport and excitatory amino acid transporters: proteins downregulated upon sample intake (p < 0.05): brain acid-soluble protein 1, microtubule-associated protein tau, synapsin-2, sodium- and chloride-dependent GABA transporter; proteins that tended to decrease upon sample intake (0.05 < p < 0.10): Parkinson’s disease (autosomal recessive and early-onset) 7 and synapsin-1; proteins upregulated upon sample intake (p > 0.95): glutathione S-transferase Mu 1, tubulin alpha-1A chain, dynamin-2, calcium/calmodulin-dependent protein kinase type II subunit gamma and tyrosine 3-monooxygenase/tyrosine 5-monooxygenase activation protein epsilon polypeptide; proteins that tended to increase upon sample intake (0.95 > p > 0.90): glutathione S-transferase Mu7 and soluble carrier family 1 (glial high-affinity glutamate transporter); proteins that tended to decrease: sodium- and chloride-dependent GABA transporter 3. These results indicate that matcha and decaffeinated matcha could reduce aging and cognitive impairment by regulating the expression of these proteins. Furthermore, these proteins could be used as markers for the evaluation of food and its available components for reducing aging and cognitive impairment.
Collapse
|
11
|
Enache D, Pereira JB, Jelic V, Winblad B, Nilsson P, Aarsland D, Bereczki E. Increased Cerebrospinal Fluid Concentration of ZnT3 Is Associated with Cognitive Impairment in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1143-1155. [PMID: 32925049 DOI: 10.3233/jad-200498] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cognitive deficits arising in the course of Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and Parkinson's disease with dementia (PDD) are directly linked to synaptic loss. Postmortem studies suggest that zinc transporter protein 3 (ZnT3), AMPA glutamate receptor 3 (GluA3), and Dynamin1 are associated with cognitive decline in AD and Lewy body dementia patients. OBJECTIVE We aimed to evaluate the diagnostic value of ZnT3, GluA3, and Dynamin 1 in the cerebrospinal fluid (CSF) of patients with dementia due to AD, DLB, and PDD compared to cognitively normal subjective cognitive decline (SCD) patients in a retrospective study. In addition, we assessed the relationship between synaptic markers and age, sex, cognitive impairment, and depressive symptoms as well as CSF amyloid, phosphorylated tau (p-tau), and total tau (T-tau). METHODS Commercially available ELISA immunoassay was used to measure the levels of proteins in a total of 97 CSF samples from AD (N = 24), PDD (N = 18), DLB (N = 27), and SCD (N = 28) patients. Cognitive impairment was assessed using the Mini-Mental State Examination (MMSE). RESULTS We found a significant increase in the concentrations of ZnT3, GluA3, and Dynamin1 in AD (p = 0.002) and of ZnT3 and Dynamin 1 in DLB (p = 0.001, p = 0.002) when compared to SCD patients. Changes in ZnT3 concentrations correlated with MMSE scores in AD (p = 0.011), and with depressive symptoms in SCD (p = 0.041). CONCLUSION We found alteration of CSF levels of synaptic proteins in AD, PDD, and DLB. Our results reveal distinct changes in CSF concentrations of ZnT3 that could reflect cognitive impairment in AD with implications for future prognostic and diagnostic marker development.
Collapse
Affiliation(s)
- Daniela Enache
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Jelic
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Dag Aarsland
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Koh YQ, Ng DQ, Ng CC, Boey A, Wei M, Sze SK, Ho HK, Acharya M, Limoli CL, Chan A. Extracellular Vesicle Proteome of Breast Cancer Patients with and Without Cognitive Impairment Following Anthracycline-based Chemotherapy: An Exploratory Study. Biomark Insights 2021; 16:11772719211018204. [PMID: 34103887 PMCID: PMC8150437 DOI: 10.1177/11772719211018204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment due to cancer and its therapy is a major concern among cancer patients and survivors. Extracellular vesicle (EVs) composition altered by cancer and chemotherapy may affect neurological processes such as neuroplasticity, potentially impacting the cognitive abilities of cancer patients and survivors. We investigated the EV proteome of breast cancer patients with and without cognitive impairment following anthracycline-based chemotherapy from longitudinally collected plasma. EVs were cup-shaped and positive for Flotillin-1 and TSG-101. We identified 517 differentially expressed EV proteins between the cognitive impaired and non-impaired groups during and post-chemotherapy. The observed decreased expression of p2X purinoceptor, cofilin-1, ADAM 10, and dynamin-1 in the plasma EVs of the cognitive impaired group may suggest alterations in the mechanisms underlying synaptic plasticity. The reduced expression of tight junction proteins among cognitive-impaired patients may imply weakening of the blood-brain barrier. These EV protein signatures may serve as a fingerprint that underscores the mechanisms underlying cognitive impairment in cancer patients and survivors.
Collapse
Affiliation(s)
- Yong Qin Koh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA, USA
| | - Chiu Chin Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Adrian Boey
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Meng Wei
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Munjal Acharya
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA, USA.,Department of Oncology Pharmacy, National Cancer Centre Singapore, Singapore
| |
Collapse
|
13
|
Arriagada-Diaz J, Prado-Vega L, Cárdenas Díaz AM, Ardiles AO, Gonzalez-Jamett AM. Dynamin Superfamily at Pre- and Postsynapses: Master Regulators of Synaptic Transmission and Plasticity in Health and Disease. Neuroscientist 2020; 28:41-58. [PMID: 33300419 DOI: 10.1177/1073858420974313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dynamin superfamily proteins (DSPs) comprise a large group of GTP-ases that orchestrate membrane fusion and fission, and cytoskeleton remodeling in different cell-types. At the central nervous system, they regulate synaptic vesicle recycling and signaling-receptor turnover, allowing the maintenance of synaptic transmission. In the presynapses, these GTP-ases control the recycling of synaptic vesicles influencing the size of the ready-releasable pool and the release of neurotransmitters from nerve terminals, whereas in the postsynapses, they are involved in AMPA-receptor trafficking to and from postsynaptic densities, supporting excitatory synaptic plasticity, and consequently learning and memory formation. In agreement with these relevant roles, an important number of neurological disorders are associated with mutations and/or dysfunction of these GTP-ases. Along the present review we discuss the importance of DSPs at synapses and their implication in different neuropathological contexts.
Collapse
Affiliation(s)
- Jorge Arriagada-Diaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Prado-Vega
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Magister en Ciencias, mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Ana M Cárdenas Díaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
14
|
Iannucci J, Johnson SL, Majchrzak M, Barlock BJ, Akhlaghi F, Seeram NP, Sen A, Grammas P. Short-term treatment with dabigatran alters protein expression patterns in a late-stage tau-based Alzheimer's disease mouse model. Biochem Biophys Rep 2020; 24:100862. [PMID: 33294639 PMCID: PMC7689047 DOI: 10.1016/j.bbrep.2020.100862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/29/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022] Open
Abstract
Proteins that regulate the coagulation cascade, including thrombin, are elevated in the brains of Alzheimer's disease (AD) patients. While studies using amyloid-based AD transgenic mouse models have implicated thrombin as a protein of interest, the role of thrombin in tau-based animal models has not been explored. The current study aims to determine how inhibiting thrombin could alter oxidative stress, inflammation, and AD-related proteins in a tau-based mouse model, the Tg4510. Aged Tg4510 mice were treated with the direct thrombin inhibitor dabigatran or vehicle for 7 days, brains collected, and western blot and data-independent proteomics using mass spectrometry with SWATH-MS acquisition performed to evaluate proteins related to oxidative stress, intracellular signaling, inflammation, and AD pathology. Dabigatran reduced iNOS, NOX4, and phosphorylation of tau (S396, S416). Additionally, dabigatran treatment increased expression of several signaling proteins related to cell survival and synaptic function. Increasing evidence supports a chronic procoagulant state in AD, highlighting a possible pathogenic role for thrombin. Our data demonstrate that inhibiting thrombin produces alterations in the expression of proteins involved in oxidative stress, inflammation, and AD-related pathology, suggesting that thrombin-mediated signaling affects multiple AD-related pathways providing a potential future therapeutic target. Thrombin inhibition with dabigatran reduces markers of oxidative stress in vivo. Dabigatran treatment reduces tau pathology in vivo. Dabigatran treatment promotes factors related to cell survival, synaptic function.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Shelby L Johnson
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.,Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Mark Majchrzak
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA
| | - Benjamin J Barlock
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Navindra P Seeram
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.,Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Abhik Sen
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA
| | - Paula Grammas
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| |
Collapse
|
15
|
Mollo N, Cicatiello R, Aurilia M, Scognamiglio R, Genesio R, Charalambous M, Paladino S, Conti A, Nitsch L, Izzo A. Targeting Mitochondrial Network Architecture in Down Syndrome and Aging. Int J Mol Sci 2020; 21:E3134. [PMID: 32365535 PMCID: PMC7247689 DOI: 10.3390/ijms21093134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are organelles that mainly control energy conversion in the cell. In addition, they also participate in many relevant activities, such as the regulation of apoptosis and calcium levels, and other metabolic tasks, all closely linked to cell viability. Functionality of mitochondria appears to depend upon their network architecture that may dynamically pass from an interconnected structure with long tubular units, to a fragmented one with short separate fragments. A decline in mitochondrial quality, which presents itself as an altered structural organization and a function of mitochondria, has been observed in Down syndrome (DS), as well as in aging and in age-related pathologies. This review provides a basic overview of mitochondrial dynamics, from fission/fusion mechanisms to mitochondrial homeostasis. Molecular mechanisms determining the disruption of the mitochondrial phenotype in DS and aging are discussed. The impaired activity of the transcriptional co-activator PGC-1α/PPARGC1A and the hyperactivation of the mammalian target of rapamycin (mTOR) kinase are emerging as molecular underlying causes of these mitochondrial alterations. It is, therefore, likely that either stimulating the PGC-1α activity or inhibiting mTOR signaling could reverse mitochondrial dysfunction. Evidence is summarized suggesting that drugs targeting either these pathways or other factors affecting the mitochondrial network may represent therapeutic approaches to improve and/or prevent the effects of altered mitochondrial function. Overall, from all these studies it emerges that the implementation of such strategies may exert protective effects in DS and age-related diseases.
Collapse
Affiliation(s)
- Nunzia Mollo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Cicatiello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Miriam Aurilia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Roberta Scognamiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Genesio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Charalambous
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Conti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Lucio Nitsch
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy
| | - Antonella Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
16
|
Park SJ, Park H, Kim MG, Zhang S, Park SE, Kim S, Chung C. Inositol Pyrophosphate Metabolism Regulates Presynaptic Vesicle Cycling at Central Synapses. iScience 2020; 23:101000. [PMID: 32252022 PMCID: PMC7132149 DOI: 10.1016/j.isci.2020.101000] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 02/17/2020] [Accepted: 03/18/2020] [Indexed: 11/27/2022] Open
Abstract
The coordination of synaptic vesicle exocytosis and endocytosis supports neurotransmitter release from presynaptic terminals. Although inositol pyrophosphates, such as 5-diphosphoinositol pentakisphosphate (5-IP7), are versatile signaling metabolites in many biological events, physiological actions of 5-IP7 on synaptic membrane vesicle trafficking remain unclear. Here, we investigated the role of 5-IP7 in synaptic transmission in hippocampal brain slices from inositol hexakisphosphate kinase 1 (Ip6k1)-knockout mice. We found that presynaptic release probability was significantly increased in Ip6k1-knockout neurons, implying enhanced activity-dependent synaptic vesicle exocytosis. Expression of wild-type but not catalytically inactive IP6K1 in the Ip6k1-knockout hippocampus restored the altered presynaptic release probability. Moreover, Ip6k1-knockout neurons were insensitive to folimycin, a vacuolar ATPase inhibitor, and dynasore, a dynamin inhibitor, suggesting marked impairment in synaptic endocytosis during exocytosis. Our findings collectively establish that IP6K1 and its product, 5-IP7, act as key physiological determinants for inhibition of presynaptic vesicle exocytosis and stimulation of endocytosis at central synapses.
Collapse
Affiliation(s)
- Seung Ju Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea
| | - Min-Gyu Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seungjae Zhang
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea
| | - Seung Eun Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; KAIST Institute for the BioCentury, KAIST, Daejeon 34141, Korea.
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
17
|
Diling C, Yinrui G, Longkai Q, Xiaocui T, Yadi L, Xin Y, Guoyan H, Ou S, Tianqiao Y, Dongdong W, Yizhen X, Yang BB, Qingping W. Circular RNA NF1-419 enhances autophagy to ameliorate senile dementia by binding Dynamin-1 and Adaptor protein 2 B1 in AD-like mice. Aging (Albany NY) 2019; 11:12002-12031. [PMID: 31860870 PMCID: PMC6949063 DOI: 10.18632/aging.102529] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022]
Abstract
Recent studies have demonstrated circular RNAs (circRNAs) to be widely expressed and to have important physiological functions. However, the expression, regulation, and function of circRNAs in neuroglial cells are unknown. Herein, we characterized the expression, regulation, and function of circRNAs in astrocytes. Astrocyte circRNAs were identified by computational analysis of newborn SD rat primary astrocytes cultured with 20 g/L D-galactose. In this manner, 7376 circRNAs were identified, among which most circRNAs (5754) were derived from annot_exons, whereas 27 were antisense, 853 were exon/intron, 329 were intergenic, 41 were intronic, and 372 were one exon. Among these, circNF1-419 was demonstrated to regulate autophagy, in over-expressing circNF1-419 transfected astrocytes, through the PI3K-I/Akt-AMPK-mTOR and PI3K-I/Akt-mTOR signaling pathways. An adenovirus associated virus packaging system (virus titer 1 ×1012), over-expressing circNF1-419 and injected into mouse cerebral cortex, showed autophagy enhancing activity by binding the proteins Dynamin-1 and Adaptor protein 2 B1 (AP2B1). This binding regulated aging markers (p21, p35/25, and p16) and inflammatory factors (TNF-α and NF-κB), and reduced the expression of Alzheimer’s disease marker proteins (Tau, p-Tau, Aβ1-42, and APOE), which delayed senile dementia. Transcriptome analysis of the brain showed that circNF1-419 improved other signaling pathways, especially those related to the synapses of SAMP8 mice. These findings provide novel insights into circNF1-419 and its potential usefulness for the diagnosis and treatment of dementia by regulating Dynamin-1 and AP2B1 mediated autophagy.
Collapse
Affiliation(s)
- Chen Diling
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Guo Yinrui
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Qi Longkai
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Tang Xiaocui
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Liu Yadi
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China.,Research and Development Institute of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yang Xin
- Department of Pharmacy, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, China
| | - Hu Guoyan
- Department of Pharmacy, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, China
| | - Shuai Ou
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yong Tianqiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Wang Dongdong
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xie Yizhen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Burton B Yang
- Sunnybrook Research Institute, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Wu Qingping
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| |
Collapse
|
18
|
Zolochevska O, Bjorklund N, Woltjer R, Wiktorowicz JE, Taglialatela G. Postsynaptic Proteome of Non-Demented Individuals with Alzheimer's Disease Neuropathology. J Alzheimers Dis 2019; 65:659-682. [PMID: 30103319 PMCID: PMC6130411 DOI: 10.3233/jad-180179] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Some individuals, here referred to as Non-Demented with Alzheimer’s Neuropathology (NDAN), retain their cognitive function despite the presence of amyloid plaques and tau tangles typical of symptomatic Alzheimer’s disease (AD). In NDAN, unlike AD, toxic amyloid-β oligomers do not localize to the postsynaptic densities (PSDs). Synaptic resistance to amyloid-β in NDAN may thus enable these individuals to remain cognitively intact despite the AD-like pathology. The mechanism(s) responsible for this resistance remains unresolved and understanding such protective biological processes could reveal novel targets for the development of effective treatments for AD. The present study uses a proteomic approach to compare the hippocampal postsynaptic densities of NDAN, AD, and healthy age-matched persons to identify protein signatures characteristic for these groups. Subcellular fractionation followed by 2D gel electrophoresis and mass spectrometry were used to analyze the PSDs. We describe fifteen proteins which comprise the unique proteomic signature of NDAN PSDs, thus setting them apart from control subjects and AD patients.
Collapse
Affiliation(s)
- Olga Zolochevska
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicole Bjorklund
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Randall Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, OR, USA
| | - John E Wiktorowicz
- Department of Biochemistry and Molecular Biology, Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Giulio Taglialatela
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
19
|
Lomash RM, Petralia RS, Holtzclaw LA, Tsuda MC, Wang YX, Badger JD, Cameron HA, Youle RJ, Roche KW. Neurolastin, a dynamin family GTPase, translocates to mitochondria upon neuronal stress and alters mitochondrial morphology in vivo. J Biol Chem 2019; 294:11498-11512. [PMID: 31177092 DOI: 10.1074/jbc.ra118.007245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/03/2019] [Indexed: 02/05/2023] Open
Abstract
Neurolastin is a dynamin family GTPase that also contains a RING domain and exhibits both GTPase and E3 ligase activities. It is specifically expressed in the brain and is important for synaptic transmission, as neurolastin knockout animals have fewer dendritic spines and exhibit a reduction in functional synapses. Our initial study of neurolastin revealed that it is membrane-associated and partially co-localizes with endosomes. Using various biochemical and cell-culture approaches, we now show that neurolastin also localizes to mitochondria in HeLa cells, cultured neurons, and brain tissue. We found that the mitochondrial localization of neurolastin depends upon an N-terminal mitochondrial targeting sequence and that neurolastin is imported into the mitochondrial intermembrane space. Although neurolastin was only partially mitochondrially localized at steady state, it displayed increased translocation to mitochondria in response to neuronal stress and mitochondrial fragmentation. Interestingly, inactivation or deletion of neurolastin's RING domain also increased its mitochondrial localization. Using EM, we observed that neurolastin knockout animals have smaller but more numerous mitochondria in cerebellar Purkinje neurons, indicating that neurolastin regulates mitochondrial morphology. We conclude that the brain-specific dynamin GTPase neurolastin exhibits stress-responsive localization to mitochondria and is required for proper mitochondrial morphology.
Collapse
Affiliation(s)
- Richa Madan Lomash
- Receptor Biology Section, NINDS, National Institutes of Health, Bethesda, Maryland 20892
| | - Ronald S Petralia
- Advanced Imaging Core, NIDCD, National Institutes of Health, Bethesda, Maryland 20892
| | - Lynne A Holtzclaw
- Microscopy and Imaging Core, NICHD, National Institutes of Health, Bethesda, Maryland 20892
| | - Mumeko C Tsuda
- Section on Neuroplasticity, NIMH, National Institutes of Health, Bethesda, Maryland 20892
| | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD, National Institutes of Health, Bethesda, Maryland 20892
| | - John D Badger
- Receptor Biology Section, NINDS, National Institutes of Health, Bethesda, Maryland 20892
| | - Heather A Cameron
- Section on Neuroplasticity, NIMH, National Institutes of Health, Bethesda, Maryland 20892
| | - Richard J Youle
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland 20892
| | - Katherine W Roche
- Receptor Biology Section, NINDS, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
20
|
Gaiardo RB, Abreu TF, Tashima AK, Telles MM, Cerutti SM. Target Proteins in the Dorsal Hippocampal Formation Sustain the Memory-Enhancing and Neuroprotective Effects of Ginkgo biloba. Front Pharmacol 2019; 9:1533. [PMID: 30666208 PMCID: PMC6330356 DOI: 10.3389/fphar.2018.01533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 12/13/2018] [Indexed: 12/13/2022] Open
Abstract
We have previously shown that standardized extracts of Ginkgo biloba (EGb) modulate fear memory formation, which is associated with CREB-1 (mRNA and protein) upregulation in the dorsal hippocampal formation (dHF), in a dose-dependent manner. Here, we employed proteomic analysis to investigate EGb effects on different protein expression patterns in the dHF, which might be involved in the regulation of CREB activity and the synaptic plasticity required for long-term memory (LTM) formation. Adult male Wistar rats were randomly assigned to four groups (n = 6/group) and were submitted to conditioned lick suppression 30 min after vehicle (12% Tween 80) or EGb (0.25, 0.50, and 1.00 g⋅kg-1) administration (p.o). All rats underwent a retention test session 48 h after conditioning. Twenty-four hours after the test session, the rats were euthanized via decapitation, and dHF samples were removed for proteome analysis using two-dimensional polyacrylamide gel electrophoresis, followed by peptide mass fingerprinting. In agreement with our previous data, no differences in the suppression ratios (SRs) were identified among the groups during first trial of CS (conditioned stimulus) presentation (P > 0.05). Acute treatment with 0.25 g⋅kg-1 EGb significantly resulted in retention of original memory, without prevent acquisition of extinction within-session. In addition, our results showed, for the first time, that 32 proteins were affected in the dHF following treatment with 0.25, 0.50, and 1.00 g⋅kg-1 doses of EGb, which upregulated seven, 19, and five proteins, respectively. Additionally, EGb downregulated two proteins at each dose. These proteins are correlated with remodeling of the cytoskeleton; the stability, size, and shape of dendritic spines; myelin sheath formation; and composition proteins of structures found in the membrane of the somatodendritic and axonal compartments. Our findings suggested that EGb modulates conditioned suppression LTM through differential protein expression profiles, which may be a target for cognitive enhancers and for the prevention or treatment of neurocognitive impairments.
Collapse
Affiliation(s)
- Renan Barretta Gaiardo
- Departamento de Ciências Biológicas, Laboratório de Farmacologia Celular e Comportamental, Universidade Federal de São Paulo, Diadema, Brazil
| | - Thiago Ferreira Abreu
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alexandre Keiji Tashima
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica Marques Telles
- Departamento de Ciências Biológicas, Laboratório de Fisiologia Metabólica, Universidade Federal de São Paulo, Diadema, Brazil
| | - Suzete Maria Cerutti
- Departamento de Ciências Biológicas, Laboratório de Farmacologia Celular e Comportamental, Universidade Federal de São Paulo, Diadema, Brazil
| |
Collapse
|
21
|
Improved synaptic and cognitive function in aged 3 × Tg-AD mice with reduced amyloid-β after immunotherapy with a novel recombinant 6Aβ15-TF chimeric vaccine. Clin Immunol 2018; 193:12-23. [PMID: 29803820 DOI: 10.1016/j.clim.2018.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/18/2018] [Accepted: 05/23/2018] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disorder impairing memory and cognition. In this study, we describe the immunogenicity and protective efficacy of the novel recombinant 6Aβ15-TF chimeric antigen as a subunit protein vaccine for AD. Recombinant 6Aβ15-TF chimeric vaccine induced strong Aβ-specific humoral immune responses without Aβ-specific T cell immunity in C57/BL6 and 3 × Tg-AD mice at different ages. As an early immunotherapy model for AD, this vaccine induced high titers of long-lasting anti-Aβ42 antibodies in aged 3 × Tg-AD mice, which led to improve behavioral performance and markedly reduced the levels of insoluble and soluble Aβ and Aβ oligomers. In agreement with these findings, immunotherapy with 6Aβ15-TF prevented the Aβ-induced decrease of presynaptic and postsynaptic proteins in aged 3 × Tg-AD mice. Our results suggest that this novel and highly immunogenic recombinant 6Aβ15-TF chimeric vaccine provides neuroprotection in AD mice and can be considered an effective AD candidate vaccine.
Collapse
|
22
|
Jensen DD, Lieu T, Halls ML, Veldhuis NA, Imlach WL, Mai QN, Poole DP, Quach T, Aurelio L, Conner J, Herenbrink CK, Barlow N, Simpson JS, Scanlon MJ, Graham B, McCluskey A, Robinson PJ, Escriou V, Nassini R, Materazzi S, Geppetti P, Hicks GA, Christie MJ, Porter CJH, Canals M, Bunnett NW. Neurokinin 1 receptor signaling in endosomes mediates sustained nociception and is a viable therapeutic target for prolonged pain relief. Sci Transl Med 2018; 9:9/392/eaal3447. [PMID: 28566424 DOI: 10.1126/scitranslmed.aal3447] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/17/2017] [Indexed: 12/25/2022]
Abstract
Typically considered to be cell surface sensors of extracellular signals, heterotrimeric GTP-binding protein (G protein)-coupled receptors (GPCRs) control many pathophysiological processes and are the target of 30% of therapeutic drugs. Activated receptors redistribute to endosomes, but researchers have yet to explore whether endosomal receptors generate signals that control complex processes in vivo and are viable therapeutic targets. We report that the substance P (SP) neurokinin 1 receptor (NK1R) signals from endosomes to induce sustained excitation of spinal neurons and pain transmission and that specific antagonism of the NK1R in endosomes with membrane-anchored drug conjugates provides more effective and sustained pain relief than conventional plasma membrane-targeted antagonists. Pharmacological and genetic disruption of clathrin, dynamin, and β-arrestin blocked SP-induced NK1R endocytosis and prevented SP-stimulated activation of cytosolic protein kinase C and nuclear extracellular signal-regulated kinase, as well as transcription. Endocytosis inhibitors prevented sustained SP-induced excitation of neurons in spinal cord slices in vitro and attenuated nociception in vivo. When conjugated to cholestanol to promote endosomal targeting, NK1R antagonists selectively inhibited endosomal signaling and sustained neuronal excitation. Cholestanol conjugation amplified and prolonged the antinociceptive actions of NK1R antagonists. These results reveal a critical role for endosomal signaling of the NK1R in the complex pathophysiology of pain and demonstrate the use of endosomally targeted GPCR antagonists.
Collapse
Affiliation(s)
- Dane D Jensen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - TinaMarie Lieu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Michelle L Halls
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Wendy L Imlach
- Discipline of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | - Quynh N Mai
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Tim Quach
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Luigi Aurelio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Joshua Conner
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Carmen Klein Herenbrink
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Nicholas Barlow
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Jamie S Simpson
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Martin J Scanlon
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Bimbil Graham
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Adam McCluskey
- School of Environmental and Life Sciences, University of Newcastle, New South Wales 2308, Australia
| | - Phillip J Robinson
- Children's Medical Research Institute, University of Sydney, New South Wales 2145, Australia
| | - Virginie Escriou
- Unité de Technologies Chimiques et Biologiques pour la Sante, CNRS UMR8258, INSERM U1022, Université Paris Descartes, Chimie ParisTech, 75006 Paris, France
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 6-50139 Florence, Italy
| | - Serena Materazzi
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 6-50139 Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, 6-50139 Florence, Italy
| | | | - Macdonald J Christie
- Discipline of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | - Christopher J H Porter
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. .,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Meritxell Canals
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. .,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. .,Australia Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Victoria 3010, Australia.,Departments of Surgery and Pharmacology, Columbia University College of Physicians and Surgeons, Columbia University, 21 Audubon Avenue, Room 209, New York City, NY 10032, USA
| |
Collapse
|
23
|
Głombik K, Stachowicz A, Trojan E, Olszanecki R, Ślusarczyk J, Suski M, Chamera K, Budziszewska B, Lasoń W, Basta-Kaim A. Evaluation of the effectiveness of chronic antidepressant drug treatments in the hippocampal mitochondria - A proteomic study in an animal model of depression. Prog Neuropsychopharmacol Biol Psychiatry 2017; 78:51-60. [PMID: 28526399 DOI: 10.1016/j.pnpbp.2017.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/31/2022]
Abstract
Several lines of evidence indicate that adverse experience in early life may be a triggering factor for disturbances in the brain mitochondrial proteins and lead to the development of depression in adulthood. On the other hand, little is known about the impact of chronic administration of various antidepressant drugs on the brain mitochondria, as a target for the pharmacotherapy of depression. The purpose of our study was to compare the impact of chronic treatment with two antidepressant drugs with different mechanisms of action, a tricyclic antidepressant (TCA), imipramine, and an antidepressant of the selective serotonin reuptake inhibitor (SSRI) class, fluoxetine, on the mitochondria-enriched subproteome profile in the hippocampus of 3-month-old male rats following a prenatal stress procedure (an animal model of depression). We clearly confirmed that chronic imipramine and fluoxetine administration not only normalized depression-like disturbances evoked by the prenatal stress procedure but also modulated the mitochondria-enriched subproteome profile in the hippocampus of adult offspring rats. In line with this, two-dimensional electrophoresis coupled with mass spectrometry showed a statistically significant down-regulation of 14-3-3 and cytochrome bc1 proteins and an up-regulation of COP9 signalosome expression after chronic imipramine treatment in the hippocampus of prenatally stressed offspring. Fluoxetine administration strongly up-regulated the expression of cathepsin D, one of the key proteins involved in the prevention of the development of neurodegenerative processes. Furthermore, this antidepressant treatment enhanced expression of proteins engaged in the improvement of learning and memory processes (STMN1, Dnm-1) as well as in mitochondrial biogenesis and defense against oxidative stress (DJ-1). These findings provide new evidence that chronic administration of antidepressants exerts a varied impact on the mitochondria-enriched subproteome in the hippocampus of adult rats following a prenatal stress procedure. In particular, the effect of fluoxetine requires additional experiments to elucidate the possible beneficial biological consequences underlying the effects mediated by this antidepressant.
Collapse
Affiliation(s)
- Katarzyna Głombik
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland
| | - Aneta Stachowicz
- Chair of Pharmacology, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Kraków, Poland
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland
| | - Rafał Olszanecki
- Chair of Pharmacology, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Kraków, Poland
| | - Joanna Ślusarczyk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland
| | - Maciej Suski
- Chair of Pharmacology, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Kraków, Poland
| | - Katarzyna Chamera
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland
| | - Bogusława Budziszewska
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland
| | - Władysław Lasoń
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343 Kraków, Poland.
| |
Collapse
|
24
|
Dynasore blocks evoked release while augmenting spontaneous synaptic transmission from primary visceral afferents. PLoS One 2017; 12:e0174915. [PMID: 28358887 PMCID: PMC5373620 DOI: 10.1371/journal.pone.0174915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/17/2017] [Indexed: 11/27/2022] Open
Abstract
The recycling of vesicle membrane fused during exocytosis is essential to maintaining neurotransmission. The GTPase dynamin is involved in pinching off membrane to complete endocytosis and can be inhibited by dynasore resulting in activity-dependent depletion of release-competent synaptic vesicles. In rat brainstem slices, we examined the effects of dynasore on three different modes of glutamate release–spontaneous, evoked, and asynchronous release–at solitary tract (ST) inputs to neurons in the nucleus of the solitary tract (NTS). Intermittent bursts of stimuli to the ST interspersed with pauses in stimulation allowed examination of these three modes in each neuron continuously. Application of 100 μM dynasore rapidly increased the spontaneous EPSC (sEPSC) frequency which was followed by inhibition of both ST-evoked EPSCs (ST-EPSC) as well as asynchronous EPSCs. The onset of ST-EPSC failures was not accompanied by amplitude reduction–a pattern more consistent with conduction block than reduced probability of vesicle release. Neither result suggested that dynasore interrupted endocytosis. The dynasore response profile resembled intense presynaptic TRPV1 activation. The TRPV1 antagonist capsazepine failed to prevent dynasore increases in sEPSC frequency but did prevent the block of the ST-EPSC. In contrast, the TRPV1 antagonist JNJ 17203212 prevented both actions of dynasore in neurons with TRPV1-expressing ST inputs. In a neuron lacking TRPV1-expressing ST inputs, however, dynasore promptly increased sEPSC rate followed by block of ST-evoked EPSCs. Together our results suggest that dynasore actions on ST-NTS transmission are TRPV1-independent and changes in glutamatergic transmission are not consistent with changes in vesicle recycling and endocytosis.
Collapse
|
25
|
Roberts JH, Liu F, Karnuta JM, Fitzgerald MC. Discovery of Age-Related Protein Folding Stability Differences in the Mouse Brain Proteome. J Proteome Res 2016; 15:4731-4741. [PMID: 27806573 DOI: 10.1021/acs.jproteome.6b00927] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Described here is the application of thermodynamic stability measurements to study age-related differences in the folding and stability of proteins in a rodent model of aging. Thermodynamic stability profiles were generated for 809 proteins in brain cell lysates from mice, aged 6 (n = 7) and 18 months (n = 9) using the Stability of Proteins from Rates of Oxidation (SPROX) technique. The biological variability of the protein stability measurements was low and within the experimental error of SPROX. A total of 83 protein hits were detected with age-related stability differences in the brain samples. Remarkably, the large majority of the brain protein hits were destabilized in the old mice, and the hits were enriched in proteins that have slow turnover rates (p < 0.07). Furthermore, 70% of the hits have been previously linked to aging or age-related diseases. These results help validate the use of thermodynamic stability measurements to capture relevant age-related proteomic changes and establish a new biophysical link between these proteins and aging.
Collapse
Affiliation(s)
- Julia H Roberts
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Fang Liu
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Jaret M Karnuta
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Michael C Fitzgerald
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| |
Collapse
|
26
|
Yoo DY, Jung HY, Kim JW, Yim HS, Kim DW, Nam H, Suh JG, Choi JH, Won MH, Yoon YS, Hwang IK. Reduction of dynamin 1 in the hippocampus of aged mice is associated with the decline in hippocampal‑dependent memory. Mol Med Rep 2016; 14:4755-4760. [PMID: 27748822 DOI: 10.3892/mmr.2016.5804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 09/07/2016] [Indexed: 11/06/2022] Open
Abstract
Dynamin 1 is a known synaptic protein, which has is key in the presynaptic regulation of endocytosis. The present study investigated the association between age and the observed changes in Morris water maze performance, and immunoreactivity and protein levels of dynamin 1 in the mouse hippocampal formation. In addition, the effects of dynasore, an inhibitor of dynamin 1, on the hippocampal dependent memory were determined to elucidate the correlation between dynamin 1 and memory. In the training phase of the Morris water maze task, the mean escape latency of the aged group (24 months old) was significantly longer, compared with that of the adult group (4 months old), although the average swimming speed and the total distance traveled during the probe trial were similar in the two groups. In the aged group, the time spent locating the target platform was significantly longer and the time spent in the correct quadrant was significantly shorter, compared with those in the adult group. In the adult group, a moderate level of dynamin 1 was detected in the hippocampal CA1 and CA3 regions, and in the dentate gyrus. In the aged group, the immunoreactivity of dynamin 1 was almost eliminated in the CA3 region and the dentate gyrus. In addition, the protein levels of dynamin 1 in the brain were significantly lower in the aged group, compared with those in the adult group. The direct infusion of dynasore, significantly reduced the contextual memory, compared with that of animals in the vehicle‑treated group. These results suggested that dynamin 1 was susceptible to the aging process, and that a reduction in dynamin 1 may result in hippocampal‑dependent memory deficits by disrupting endocytosis and the release of neurotransmitters.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Sun Yim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Hajin Nam
- Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jun Gyo Suh
- Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
27
|
A novel recombinant 6Aβ15-THc-C chimeric vaccine (rCV02) mitigates Alzheimer's disease-like pathology, cognitive decline and synaptic loss in aged 3 × Tg-AD mice. Sci Rep 2016; 6:27175. [PMID: 27255752 PMCID: PMC4891678 DOI: 10.1038/srep27175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that impairs memory and cognition. Targeting amyloid-β (Aβ) may be currently the most promising immunotherapeutic strategy for AD. In this study, a recombinant chimeric 6Aβ15-THc-C immunogen was formulated with alum adjuvant as a novel Aβ B-cell epitope candidate vaccine (rCV02) for AD. We examined its efficacy in preventing the cognitive deficit and synaptic impairment in 3 × Tg-AD mice. Using a toxin-derived carrier protein, the rCV02 vaccine elicited robust Aβ-specific antibodies that markedly reduced AD-like pathology and improved behavioral performance in 3 × Tg-AD mice. Along with the behavioral improvement in aged 3 × Tg-AD mice, rCV02 significantly decreased calpain activation concurrent with reduced soluble Aβ or oligomeric forms of Aβ, probably by preventing dynamin 1 and PSD-95 degradation. Our data support the hypothesis that reducing Aβ levels in rCV02-immunized AD mice increases the levels of presynaptic dynamin 1 and postsynaptic PSD-95 allowing functional recovery of cognition. In conclusion, this novel and highly immunogenic rCV02 shows promise as a new candidate prophylactic vaccine for AD and may be useful for generating rapid and strong Aβ-specific antibodies in AD patients with pre-existing memory Th cells generated after immunization with conventional tetanus toxoid vaccine.
Collapse
|
28
|
A Novel Aβ B-Cell Epitope Vaccine (rCV01) for Alzheimer's Disease Improved Synaptic and Cognitive Functions in 3 × Tg-AD Mice. J Neuroimmune Pharmacol 2016; 11:657-668. [PMID: 27147259 DOI: 10.1007/s11481-016-9678-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive amyloid-β accumulation, loss of cognitive abilities, and synaptic alterations. Given the remarkable recovery of cognition in AD models of targeting-Aβ immunotherapy, we sought to determine the molecular correlate(s) associated with improvement. We evaluated the efficacy of a recombinant chimeric 6Aβ15-T antigen formulated with alum adjuvant as a novel Aβ B-cell epitope vaccine (rCV01) in 3 × Tg-AD mice. rCV01 elicited robust Th2-polarized Aβ-specific antibodies without autoimmune T cell responses in 3 × Tg-AD mice. The long-lasting anti-Aβ42 antibodies were associated with markedly reduced AD-like pathology, enhanced synaptic function, and improved cognitive performance in aged 3 × Tg-AD mice. This is the first report to provide one hypothesis for the improved outcomes following vaccination is a reduction in the levels of active calpain in rCV01-immunized AD mice, which is likely attributable to preventing dynamin 1 and PSD-95 degradation allowing functional recovery of cognition. rCV01 is a highly immunogenic recombinant chimeric 6Aβ15-T vaccine that shows clear neuroprotective properties in preclinical mouse models of AD and is a candidate for an effective AD vaccine.
Collapse
|
29
|
Peripherally administered sera antibodies recognizing amyloid-β oligomers mitigate Alzheimer's disease-like pathology and cognitive decline in aged 3× Tg-AD mice. Vaccine 2016; 34:1758-66. [PMID: 26945100 DOI: 10.1016/j.vaccine.2016.02.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/05/2016] [Accepted: 02/19/2016] [Indexed: 02/06/2023]
Abstract
Active and passive immunotherapy targeting amyloid-β (Aβ) may be the most promising strategy to prevent or treat Alzheimer's disease (AD). Previously, immunization with the recombinant 6Aβ15-T antigen generated robust anti-Aβ serum antibodies that strongly recognized Aβ42 oligomers in different mice, markedly reduced the amyloid burden, and improved behavioral performance of immunized older AD mice. Here, we further determined that these anti-6Aβ15-T serum antibodies from different strains of mice displayed anti-Aβ antibody responses against the same epitopes in the Aβ1-15 region. Peripheral administration of anti-6Aβ15-T serum antibodies was also effective to mitigate AD-like pathology and cognitive decline in aged 3× Tg-AD mice. Specifically, the levels of Aβ and tau in the brains of 3× Tg-AD mice were significantly reduced after passive immunotherapy, which seemed necessary or beneficial to ameliorate memory impairment. In addition, our results showed that this immunotherapy also prevented presynaptic dynamin 1 degradation, which might help to further protect synaptic functions and allow functional recovery of cognition. Moreover, immunization with 6Aβ15-T in rabbits induced a similar antibody response as that in mice, and the rabbit serum antibodies reacted strongly with Aβ42 oligomers and inhibited oligomer-mediated neurotoxicity. We concluded that passive immunization with Aβ42 oligomer conformation-sensitive anti-6Aβ15-T serum antibodies is effective in providing potentially therapeutic effects in aged 3× Tg-AD mice by reducing Aβ and tau.
Collapse
|
30
|
Fà M, Zhang H, Staniszewski A, Saeed F, Shen LW, Schiefer IT, Siklos MI, Tapadar S, Litosh VA, Libien J, Petukhov PA, Teich AF, Thatcher GR, Arancio O. Novel Selective Calpain 1 Inhibitors as Potential Therapeutics in Alzheimer's Disease. J Alzheimers Dis 2016; 49:707-21. [PMID: 26484927 PMCID: PMC8962836 DOI: 10.3233/jad-150618] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease, one of the most important brain pathologies associated with neurodegenerative processes, is related to overactivation of calpain-mediated proteolysis. Previous data showed a compelling efficacy of calpain inhibition against abnormal synaptic plasticity and memory produced by the excess of amyloid-β, a distinctive marker of the disease. Moreover, a beneficial effect of calpain inhibitors in Alzheimer's disease is predictable by the occurrence of calpain hyperactivation leading to impairment of memory-related pathways following abnormal calcium influxes that might ensue independently of amyloid-β elevation. However, molecules currently available as effective calpain inhibitors lack adequate selectivity. This work is aimed at characterizing the efficacy of a novel class of epoxide-based inhibitors, synthesized to display improved selectivity and potency towards calpain 1 compared to the prototype epoxide-based generic calpain inhibitor E64. Both functional and preliminary toxicological investigations proved the efficacy, potency, and safety of the novel and selective calpain inhibitors NYC438 and NYC488 as possible therapeutics against the disease.
Collapse
Affiliation(s)
- Mauro Fà
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Hong Zhang
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Agnieszka Staniszewski
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Faisal Saeed
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Li W. Shen
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Isaac T. Schiefer
- Department of Medicinal and Biological Chemistry, University of Ohio at Toledo, Frederic and Mary Wolfe Center, Toledo, OH, USA
| | - Marton I. Siklos
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Subhasish Tapadar
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Vladislav A. Litosh
- Department of Chemistry, McMicken College of Arts & Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Jenny Libien
- Department of Pathology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Pavel A. Petukhov
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew F. Teich
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Gregory R.J. Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| |
Collapse
|
31
|
Borovok N, Nesher E, Levin Y, Reichenstein M, Pinhasov A, Michaelevski I. Dynamics of Hippocampal Protein Expression During Long-term Spatial Memory Formation. Mol Cell Proteomics 2015; 15:523-41. [PMID: 26598641 DOI: 10.1074/mcp.m115.051318] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Indexed: 01/08/2023] Open
Abstract
Spatial memory depends on the hippocampus, which is particularly vulnerable to aging. This vulnerability has implications for the impairment of navigation capacities in older people, who may show a marked drop in performance of spatial tasks with advancing age. Contemporary understanding of long-term memory formation relies on molecular mechanisms underlying long-term synaptic plasticity. With memory acquisition, activity-dependent changes occurring in synapses initiate multiple signal transduction pathways enhancing protein turnover. This enhancement facilitates de novo synthesis of plasticity related proteins, crucial factors for establishing persistent long-term synaptic plasticity and forming memory engrams. Extensive studies have been performed to elucidate molecular mechanisms of memory traces formation; however, the identity of plasticity related proteins is still evasive. In this study, we investigated protein turnover in mouse hippocampus during long-term spatial memory formation using the reference memory version of radial arm maze (RAM) paradigm. We identified 1592 proteins, which exhibited a complex picture of expression changes during spatial memory formation. Variable linear decomposition reduced significantly data dimensionality and enriched three principal factors responsible for variance of memory-related protein levels at (1) the initial phase of memory acquisition (165 proteins), (2) during the steep learning improvement (148 proteins), and (3) the final phase of the learning curve (123 proteins). Gene ontology and signaling pathways analysis revealed a clear correlation between memory improvement and learning phase-curbed expression profiles of proteins belonging to specific functional categories. We found differential enrichment of (1) neurotrophic factors signaling pathways, proteins regulating synaptic transmission, and actin microfilament during the first day of the learning curve; (2) transcription and translation machinery, protein trafficking, enhancement of metabolic activity, and Wnt signaling pathway during the steep phase of memory formation; and (3) cytoskeleton organization proteins. Taken together, this study clearly demonstrates dynamic assembly and disassembly of protein-protein interaction networks depending on the stage of memory formation engrams.
Collapse
Affiliation(s)
- Natalia Borovok
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Elimelech Nesher
- §Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Yishai Levin
- ¶de Botton Institute for Protein Profiling, The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Reichenstein
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Albert Pinhasov
- §Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Izhak Michaelevski
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel; ‖Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
32
|
Dempwolff F, Graumann PL. Genetic links between bacterial dynamin and flotillin proteins. Commun Integr Biol 2014; 7:970972. [PMID: 26842743 PMCID: PMC4594503 DOI: 10.4161/cib.29578] [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: 05/13/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 01/29/2023] Open
Abstract
Dynamin is a membrane-associated GTPase that confers motor-like functions in membrane
dynamics, such as endocytosis, in eukaryotic cells. Flotillin (reggie) proteins are also a
widely conserved class of membrane proteins, associated with the formation of protein
assemblies within the membrane, and with endocytotic processes. Bacterial dynamin has been
shown to bind to membranes in vitro and to mediate membrane fusion. Bacillus
subtilis DynA localizes to the cell division septum, and it was recently shown
that it indeed plays a role in cell division. Interestingly, dynamin shows a genetic
interaction with flotillin proteins in this prokaryotic model organism and the absence of
both proteins results in a cell division and cell shape defect. Here, we show that in
addition to the morphological phenotypes, a dynamin/flotillin double deletion strain shows
a synthetic defect in cell motility, much stronger than that of flotillin single mutant
cells. While the contribution of altered cell shape and slower growth of the double
deletion strain on motility cannot be clearly assessed, our data emphasize the fact that
dynamin and flotillin proteins play tightly connected functions in a wide range of aspects
in membrane processes in bacteria.
Collapse
Affiliation(s)
- Felix Dempwolff
- SYNMIKRO; LOEWE-Centre for Synthetic Microbiology ; Marburg, Germany
| | - Peter L Graumann
- SYNMIKRO; LOEWE-Centre for Synthetic Microbiology; Department of Chemistry ; Marburg, Germany
| |
Collapse
|