1
|
Attia GM, Ali LS, Eldesoqui M, Elsaed WM, Mostafa SA, Albadawi EA, Elmansy RA, Elhassan YH, Berika M, Badawy AA, El-Nabalaway M, Dawood AF, Seleem HS. Neuroprotective effects of granulocyte colony-stimulating factor against tramadol-induced cerebellar neurotoxicity. Tissue Cell 2025; 94:102832. [PMID: 40048827 DOI: 10.1016/j.tice.2025.102832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Tramadol (TRM) is a centrally acting synthetic opioid and serotonin/norepinephrine reuptake inhibitor. Despite being a potent painkiller, long-term use can induce permanent neurotoxicity. Granulocyte colony-stimulating factor (G-CSF) is a cytokine that helps to mobilize stem cells and facilitate their integration over injured neurons. AIM This work aims to study the histopathological, biochemical, and molecular alterations in the cerebellar cortex induced by TRM in comparison to the postulated protective effect of G-CSF versus TRM withdrawal. METHODS 32 adult male albino rats were equally divided into four groups: control, TRM, TRM+G-CSF-treated, and TRM withdrawal groups. The TRM group received a daily dose of 80 mg/kg body weight orally via gastric tube for 12 weeks. The TRM+G-CSF-treated group received subcutaneous injections of 100 μg/kg body weight of G-CSF for seven consecutive days, then TRM from the 8th day. The TRM withdrawal group received TRM for 12 weeks; then, the rats were left without TRM administration for a further 12 weeks. The structural, biochemical, and molecular changes of the cerebellum were measured. RESULTS The study revealed that TRM not only induced cerebellar atrophy but also triggered microgliosis, neuroinflammation, and apoptotic indicators, all while suppressing autophagy. However, G-CSF and TRM withdrawal reversed these alterations with superiority to G-CSF. CONCLUSION The current investigation shows that G-CSF may improve behavioral, neurochemical, immunohistochemical, and molecular metrics in the rat cerebellum after tramadol-induced injury. G-CSF exhibits a superior protective effect compared to tramadol withdrawal. This is achieved through its antioxidant, anti-apoptotic, and autophagic enhancement properties, as well as its ability to reduce cerebellar gliosis.
Collapse
Affiliation(s)
- Ghalia Mahfouz Attia
- Department of Medical Histology and Cell Biology, Faculty of Medicine Mansoura University, Egypt; Department of Medical Histology and Cell Biology, Faculty of Medicine Horus University, Egypt.
| | - Lashin S Ali
- Department of Basic Medical Science-Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan; Physiology Department-Mansoura Faculty of Medine-Mansoura University, Mansoura, Egypt.
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia.
| | - Wael M Elsaed
- Department of Human Anatomy and Embryology, Faculty of Medicine Mansoura University, Egypt; Basic Sciences Department, Riyadh Elm University, Riyadh, Saudi Arabia.
| | - Sally Abdallah Mostafa
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Mansoura University, Egypt.
| | - Emad A Albadawi
- Department of Basic Medical Sciences, College of Medicine, Taibah University, KSA.
| | - Rasha Ahmed Elmansy
- Anatomy Unit, Department of Basic Medical Sciences, College of Medicine, Qassim University, Buraydah, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | | | - Mohamed Berika
- Department of Human Anatomy and Embryology, Faculty of Medicine Mansoura University, Egypt; Rehabilitation Science Department, College of Applied Medical Sciences, King Saud University, KSA.
| | - Abdelnaser A Badawy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia.
| | - Mohammad El-Nabalaway
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine Mansoura University, Egypt.
| | - Amal Fahmy Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hanan Said Seleem
- Department of Histology & Cell Biology, Faculty of Medicine, Menoufia University, Shebin ElKoum, Menofia, Egypt.
| |
Collapse
|
2
|
Ejaz S, Sternburg JO, Rezvani K, Ahammed MS, Giri S, Liu J, Wang H, Wang X. Ser14-phosphorylated Rpn6 Limits Proteostasis Impairment and Pathology in Both Brain and Heart of Tauopathy Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645024. [PMID: 40196506 PMCID: PMC11974871 DOI: 10.1101/2025.03.24.645024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Alzheimer's disease (AD) patients often display neurobehavioral and cardiac impairments, but the underlying factors remain unclear. Ser14 phosphorylation in RPN6 (p-S14-RPN6) mediates the activation of 26S proteasomes by protein kinase A (PKA). Proteasome priming is implicated in protection by cAMP-PKA against AD, but this remains to be established. Hence, this study was conducted to interrogate homeostatic p-S14-RPN6 in AD. The recently validated Rpn6 S14A knock-in (S14A) mice were crossbred with the PS19 tauopathy mice (RRID: IMSR_JAX:008169). The resultant wild type (WT), PS19, and PS19::S14A littermates were compared. Expedited declines in cognitive and motor functions as indicated respectively by significant decreases in object recognition and discrimination indexes and rotarod time were observed in PS19::S14A mice vs. PS19 mice, which is associated with more pronounced synaptic losses, microglial activation, and gliosis in the hippocampus. Compared with WT and PS19 mice, PS19::S14A mice showed exacerbated cardiac malfunction, cardiac hypertrophic responses and fibrosis, and greater increases of total and hyperphosphorylated tau proteins and ubiquitin conjugates in both hippocampi and hearts. These findings demonstrate that genetic blockade of p-S14-RPN6 exacerbates tauopathy in both the brain and heart, which for the first time establishes that homeostatic p-S14-RPN6 promotes proteostasis and protects against pathogenesis in AD.
Collapse
|
3
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
4
|
Bej E, Cesare P, d’Angelo M, Volpe AR, Castelli V. Neuronal Cell Rearrangement During Aging: Antioxidant Compounds as a Potential Therapeutic Approach. Cells 2024; 13:1945. [PMID: 39682694 PMCID: PMC11639796 DOI: 10.3390/cells13231945] [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: 10/17/2024] [Revised: 11/02/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Aging is a natural process that leads to time-related changes and a decrease in cognitive abilities, executive functions, and attention. In neuronal aging, brain cells struggle to respond to oxidative stress. The structure, function, and survival of neurons can be mediated by different pathways that are sensitive to oxidative stress and age-related low-energy states. Mitochondrial impairment is one of the most noticeable signs of brain aging. Damaged mitochondria are thought to be one of the main causes that feed the inflammation related to aging. Also, protein turnover is involved in age-related impairments. The brain, due to its high oxygen usage, is particularly susceptible to oxidative damage. This review explores the mechanisms underlying neuronal cell rearrangement during aging, focusing on morphological changes that contribute to cognitive decline and increased susceptibility to neurodegenerative diseases. Potential therapeutic approaches are discussed, including the use of antioxidants (e.g., Vitamin C, Vitamin E, glutathione, carotenoids, quercetin, resveratrol, and curcumin) to mitigate oxidative damage, enhance mitochondrial function, and maintain protein homeostasis. This comprehensive overview aims to provide insights into the cellular and molecular processes of neuronal aging and highlight promising therapeutic avenues to counteract age-related neuronal deterioration.
Collapse
Affiliation(s)
- Erjola Bej
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
- Department of the Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| | - Patrizia Cesare
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
| | - Anna Rita Volpe
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.B.); (P.C.); (M.d.)
- Department of the Chemical-Toxicological and Pharmacological Evaluation of Drugs, Faculty of Pharmacy, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| |
Collapse
|
5
|
Valdes PA, Yu CC(J, Aronson J, Ghosh D, Zhao Y, An B, Bernstock JD, Bhere D, Felicella MM, Viapiano MS, Shah K, Chiocca EA, Boyden ES. Improved immunostaining of nanostructures and cells in human brain specimens through expansion-mediated protein decrowding. Sci Transl Med 2024; 16:eabo0049. [PMID: 38295184 PMCID: PMC10911838 DOI: 10.1126/scitranslmed.abo0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Proteins are densely packed in cells and tissues, where they form complex nanostructures. Expansion microscopy (ExM) variants have been used to separate proteins from each other in preserved biospecimens, improving antibody access to epitopes. Here, we present an ExM variant, decrowding expansion pathology (dExPath), that can expand proteins away from each other in human brain pathology specimens, including formalin-fixed paraffin-embedded (FFPE) clinical specimens. Immunostaining of dExPath-expanded specimens reveals, with nanoscale precision, previously unobserved cellular structures, as well as more continuous patterns of staining. This enhanced molecular staining results in observation of previously invisible disease marker-positive cell populations in human glioma specimens, with potential implications for tumor aggressiveness. dExPath results in improved fluorescence signals even as it eliminates lipofuscin-associated autofluorescence. Thus, this form of expansion-mediated protein decrowding may, through improved epitope access for antibodies, render immunohistochemistry more powerful in clinical science and, perhaps, diagnosis.
Collapse
Affiliation(s)
- Pablo A. Valdes
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, 77555
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA, 02115
- Media Arts and Sciences, MIT, Cambridge, MA, USA, 02115
| | - Chih-Chieh (Jay) Yu
- Media Arts and Sciences, MIT, Cambridge, MA, USA, 02115
- Department of Biological Engineering, MIT, MA, USA, 02139
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA, 02139
- RIKEN Center for Brain Science, Saitama, Japan, 351-0198
| | - Jenna Aronson
- Media Arts and Sciences, MIT, Cambridge, MA, USA, 02115
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA, 02139
- RIKEN Center for Brain Science, Saitama, Japan, 351-0198
| | - Debarati Ghosh
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA, 02139
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA, 02139
| | - Yongxin Zhao
- Media Arts and Sciences, MIT, Cambridge, MA, USA, 02115
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA, 15213
| | - Bobae An
- Media Arts and Sciences, MIT, Cambridge, MA, USA, 02115
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA, 02139
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA, 02115
- Koch Institute, MIT, Cambridge, MA, USA, 02139
| | - Deepak Bhere
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA, 02115
- Department of Pathology, Microbiology and Immunology, School of Medicine Columbia, University of South Carolina, Columbia, SC, USA, 29209
- Center for Stem Cell and Translational Immunotherapy, Harvard Medical School/Brigham and Women’s Hospital, Boston, MA, USA, 02115
| | - Michelle M. Felicella
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA, 77555
| | - Mariano S. Viapiano
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA, 13210
| | - Khalid Shah
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA, 02115
- Center for Stem Cell and Translational Immunotherapy, Harvard Medical School/Brigham and Women’s Hospital, Boston, MA, USA, 02115
| | - E. Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA, 02115
| | - Edward S. Boyden
- Media Arts and Sciences, MIT, Cambridge, MA, USA, 02115
- Department of Biological Engineering, MIT, MA, USA, 02139
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA, 02139
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA, 02139
- Koch Institute, MIT, Cambridge, MA, USA, 02139
- MIT Center for Neurobiological Engineering and K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, USA, 02139
- Howard Hughes Medical Institute, Cambridge, MA, USA, 02139
| |
Collapse
|
6
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
7
|
Chen K, Garcia Padilla C, Kiselyov K, Kozai TDY. Cell-specific alterations in autophagy-lysosomal activity near the chronically implanted microelectrodes. Biomaterials 2023; 302:122316. [PMID: 37738741 PMCID: PMC10897938 DOI: 10.1016/j.biomaterials.2023.122316] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/22/2023] [Accepted: 09/02/2023] [Indexed: 09/24/2023]
Abstract
Intracortical microelectrodes that can record and stimulate brain activity have become a valuable technique for basic science research and clinical applications. However, long-term implantation of these microelectrodes can lead to progressive neurodegeneration in the surrounding microenvironment, characterized by elevation in disease-associated markers. Dysregulation of autophagy-lysosomal degradation, a major intracellular waste removal process, is considered a key factor in the onset and progression of neurodegenerative diseases. It is plausible that similar dysfunctions in autophagy-lysosomal degradation contribute to tissue degeneration following implantation-induced focal brain injury, ultimately impacting recording performance. To understand how the focal, persistent brain injury caused by long-term microelectrode implantation impairs autophagy-lysosomal pathway, we employed two-photon microscopy and immunohistology. This investigation focused on the spatiotemporal characterization of autophagy-lysosomal activity near the chronically implanted microelectrode. We observed an aberrant accumulation of immature autophagy vesicles near the microelectrode over the chronic implantation period. Additionally, we found deficits in autophagy-lysosomal clearance proximal to the chronic implant, which was associated with an accumulation of autophagy cargo and a reduction in lysosomal protease level during the chronic period. Furthermore, our evidence demonstrates reactive astrocytes have myelin-containing lysosomes near the microelectrode, suggesting its role of myelin engulfment during acute implantation period. Together, this study sheds light on the process of brain tissue degeneration caused by long-term microelectrode implantation, with a specific focus on impaired intracellular waste degradation.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Camila Garcia Padilla
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Wellman SM, Coyne OA, Douglas MM, Kozai TDY. Aberrant accumulation of age- and disease-associated factors following neural probe implantation in a mouse model of Alzheimer's disease. J Neural Eng 2023; 20:046044. [PMID: 37531953 PMCID: PMC10594264 DOI: 10.1088/1741-2552/aceca5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/04/2023]
Abstract
Objective. Electrical stimulation has had a profound impact on our current understanding of nervous system physiology and provided viable clinical options for addressing neurological dysfunction within the brain. Unfortunately, the brain's immune suppression of indwelling microelectrodes currently presents a major roadblock in the long-term application of neural recording and stimulating devices. In some ways, brain trauma induced by penetrating microelectrodes produces similar neuropathology as debilitating brain diseases, such as Alzheimer's disease (AD), while also suffering from end-stage neuron loss and tissue degeneration. The goal of the present study was to understand whether there may be any parallel mechanisms at play between brain injury from chronic microelectrode implantation and those of neurodegenerative disorder.Approach. We used two-photon microscopy to visualize the accumulation, if any, of age- and disease-associated factors around chronically implanted electrodes in both young and aged mouse models of AD.Main results. We determined that electrode injury leads to aberrant accumulation of lipofuscin, an age-related pigment, in wild-type and AD mice alike. Furthermore, we reveal that chronic microelectrode implantation reduces the growth of pre-existing Alzheimer's plaques while simultaneously elevating amyloid burden at the electrode-tissue interface. Lastly, we uncover novel spatial and temporal patterns of glial reactivity, axonal and myelin pathology, and neurodegeneration related to neurodegenerative disease around chronically implanted microelectrodes.Significance. This study offers multiple novel perspectives on the possible neurodegenerative mechanisms afflicting chronic brain implants, spurring new potential avenues of neuroscience investigation and design of more targeted therapies for improving neural device biocompatibility and treatment of degenerative brain disease.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Olivia A Coyne
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Madeline M Douglas
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
9
|
Viegas MPC, Santos LEC, Aarão MC, Cecilio SG, Medrado JM, Pires AC, Rodrigues AM, Scorza CA, Moret MA, Finsterer J, Scorza FA, Almeida ACG. The nonsynaptic plasticity in Parkinson's disease: Insights from an animal model. Clinics (Sao Paulo) 2023; 78:100242. [PMID: 37480642 PMCID: PMC10387572 DOI: 10.1016/j.clinsp.2023.100242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND The 6-OHDA nigro-striatal lesion model has already been related to disorders in the excitability and synchronicity of neural networks and variation in the expression of transmembrane proteins that control intra and extracellular ionic concentrations, such as cation-chloride cotransporters (NKCC1 and KCC2) and Na+/K+-ATPase and, also, to the glial proliferation after injury. All these non-synaptic mechanisms have already been related to neuronal injury and hyper-synchronism processes. OBJECTIVE The main objective of this study is to verify whether mechanisms not directly related to synaptic neurotransmission could be involved in the modulation of nigrostriatal pathways. METHODS Male Wistar rats, 3 months old, were submitted to a unilateral injection of 24 µg of 6-OHDA, in the striatum (n = 8). The animals in the Control group (n = 8) were submitted to the same protocol, with the replacement of 6-OHDA by 0.9% saline. The analysis by optical densitometry was performed to quantify the immunoreactivity intensity of GFAP, NKCC1, KCC2, Na+/K+-ATPase, TH and Cx36. RESULTS The 6-OHDA induced lesions in the striatum, were not followed by changes in the expression cation-chloride cotransporters and Na+/K+-ATPase, but with astrocytic reactivity in the lesioned and adjacent regions of the nigrostriatal. Moreover, the dopaminergic degeneration caused by 6-OHDA is followed by changes in the expression of connexin-36. CONCLUSIONS The use of the GJ blockers directly along the nigrostriatal pathways to control PD motor symptoms is conjectured. Electrophysiology of the striatum and the substantia nigra, to verify changes in neuronal synchronism, comparing brain slices of control animals and experimental models of PD, is needed.
Collapse
Affiliation(s)
- Mônica P C Viegas
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Luiz E C Santos
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Mayra C Aarão
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Samyra G Cecilio
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Joana M Medrado
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Arthur C Pires
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Antônio M Rodrigues
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil
| | - Carla A Scorza
- Neuroscience Discipline, Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil; Centro de Neurociências e Saúde da Mulher "Professor Geraldo Rodrigues de Lima", Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Marcelo A Moret
- SENAI ‒ Departamento Regional da Bahia, Centro Integrado de Manufatura e Tecnologia, Bahia, BA, Brazil
| | | | - Fulvio A Scorza
- Neuroscience Discipline, Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil; Centro de Neurociências e Saúde da Mulher "Professor Geraldo Rodrigues de Lima", Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil.
| | - Antônio-Carlos G Almeida
- Laboratory of Experimental and Computational Neuroscience, Department of Biosystems Engineering, Universidade Federal de São João del-Rei (UFSJ), São João del-Rei, MG, Brazil; Centro de Neurociências e Saúde da Mulher "Professor Geraldo Rodrigues de Lima", Escola Paulista de Medicina da Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
10
|
Wellman SM, Coyne OA, Douglas MM, Kozai TDY. Aberrant accumulation of age- and disease-associated factors following neural probe implantation in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.11.528131. [PMID: 36891286 PMCID: PMC9993955 DOI: 10.1101/2023.02.11.528131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Electrical stimulation has had a profound impact on our current understanding of nervous system physiology and provided viable clinical options for addressing neurological dysfunction within the brain. Unfortunately, the brain's immune suppression of indwelling microelectrodes currently presents a major roadblock in the long-term application of neural recording and stimulating devices. In some ways, brain trauma induced by penetrating microelectrodes produces similar neuropathology as debilitating brain diseases, such as Alzheimer's disease (AD), while also suffering from end-stage neuron loss and tissue degeneration. To understand whether there may be any parallel mechanisms at play between brain injury from chronic microelectrode implantation and those of neurodegenerative disorder, we used two-photon microscopy to visualize the accumulation, if any, of age- and disease-associated factors around chronically implanted electrodes in both young and aged mouse models of AD. With this approach, we determined that electrode injury leads to aberrant accumulation of lipofuscin, an age-related pigment, in wild-type and AD mice alike. Furthermore, we reveal that chronic microelectrode implantation reduces the growth of pre-existing amyloid plaques while simultaneously elevating amyloid burden at the electrode-tissue interface. Lastly, we uncover novel spatial and temporal patterns of glial reactivity, axonal and myelin pathology, and neurodegeneration related to neurodegenerative disease around chronically implanted microelectrodes. This study offers multiple novel perspectives on the possible neurodegenerative mechanisms afflicting chronic brain implants, spurring new potential avenues of neuroscience investigation and design of more targeted therapies for improving neural device biocompatibility and treatment of degenerative brain disease.
Collapse
|
11
|
Yun Y, Wang X, Xu J, Jin C, Chen J, Wang X, Wang J, Qin L, Yang P. Pristane induced lupus mice as a model for neuropsychiatric lupus (NPSLE). BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:3. [PMID: 36765366 PMCID: PMC9921421 DOI: 10.1186/s12993-023-00205-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/13/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND The pristane-induced lupus (PIL) model is a useful tool for studying environmental-related systemic lupus erythematosus (SLE). However, neuropsychiatric manifestations in this model have not been investigated in detail. Because neuropsychiatric lupus (NPSLE) is an important complication of SLE, we investigated the neuropsychiatric symptoms in the PIL mouse model to evaluate its suitability for NPSLE studies. RESULTS PIL mice showed olfactory dysfunction accompanied by an anxiety- and depression-like phenotype at month 2 or 4 after pristane injection. The levels of cytokines (IL-1β, IFN-α, IFN-β, IL-10, IFN-γ, IL-6, TNF-α and IL-17A) and chemokines (CCL2 and CXCL10) in the brain and blood-brain barrier (BBB) permeability increased significantly from week 2 or month 1, and persisted throughout the observed course of the disease. Notably, IgG deposition in the choroid plexus and lateral ventricle wall were observed at month 1 and both astrocytes and microglia were activated. Persistent activation of astrocytes was detected throughout the observed course of the disease, while microglial activation diminished dramatically at month 4. Lipofuscin deposition, a sign of neuronal damage, was detected in cortical and hippocampal neurons from month 4 to 8. CONCLUSION PIL mice exhibit a series of characteristic behavioral deficits and pathological changes in the brain, and therefore might be suitable for investigating disease pathogenesis and for evaluating potential therapeutic targets for environmental-related NPSLE.
Collapse
Affiliation(s)
- Yang Yun
- grid.412467.20000 0004 1806 3501Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuejiao Wang
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Jingyi Xu
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chenye Jin
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jingyu Chen
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Xueru Wang
- grid.412449.e0000 0000 9678 1884Department of Physiology, China Medical University, Shenyang, China
| | - Jianing Wang
- grid.412636.40000 0004 1757 9485Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang, China.
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
12
|
Naguib AM, Apparoo Y, Xiong C, Phan CW. Maitake Medicinal Mushroom, Grifola frondosa (Agaricomycetes), and Its Neurotrophic Properties: A Mini-Review. Int J Med Mushrooms 2023; 25:11-22. [PMID: 36749053 DOI: 10.1615/intjmedmushrooms.2022046849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neurodegeneration is one of the most common manifestations in an aging population. The occurrence of oxidative stress and neuroinflammation are the main contributors to the phenomenon. Neurologic conditions such as Alzheimer's disease (AD) and Parkinson's disease (PD) are challenging to treat due to their irreversible manner as well as the lack of effective treatment. Grifola frondosa (Dicks.: Fr.) S.F. Gray, or maitake mushroom, is believed to be a potential choice as a therapeutic agent for neurodegenerative diseases. G. frondosa is known to be a functional food that has a wide variety of medicinal purposes. Thus, this review emphasizes the neuroprotective effects and the chemical composition of G. frondosa. Various studies have described that G. frondosa can protect and proliferate neuronal cells through neurogenesis, antioxidative, anti-inflammatory, and anti-β-amyloid activities. The mechanism of action behind these therapeutic findings in various in vitro and in vivo models has also been intensively studied. In this mini review, we also summarized the chemical composition of G. frondosa to provide a better understanding of the presence of nutritional compounds in G. frondosa.
Collapse
Affiliation(s)
- Adrina Mohamad Naguib
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yasaaswini Apparoo
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Mushroom Research Centre, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chuan Xiong
- Biotechnology and Nuclear Technology Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 610061, P.R. China
| | - Chia Wei Phan
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Mushroom Research Centre, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Clinical Investigation Centre, 5th Floor, East Tower, University Malaya Medical Centre, 59100 Lembah Pantai Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Bandaru LJM, Murumulla L, C BL, D KP, Challa S. Exposure of combination of environmental pollutant, lead (Pb) and β-amyloid peptides causes mitochondrial dysfunction and oxidative stress in human neuronal cells. J Bioenerg Biomembr 2023; 55:79-89. [PMID: 36637735 DOI: 10.1007/s10863-023-09956-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023]
Abstract
Exposure to the environmental pollutant lead (Pb) has been linked to Alzheimer's disease (AD), in which mitochondrial dysfunction is a pathological consequence of neuronal degeneration. The toxicity of Pb in combination with β-amyloid peptides (1-40) and (25-35) causes selective death in neuronal cells. However, the precise mechanism through which Pb induces Alzheimer's disease, particularly mitochondrial damage, is unknown. Changes in mitochondrial mass, membrane potential, mitochondrial complex activities, mitochondrial DNA and oxidative stress were examined in neuronal cells of human origin exposed to Pb and β-amyloid peptides (1-40) and (25-35) individually and in different combinations. The results showed depolarization of mitochondrial membrane potential, decrease in mitochondrial mass, ATP levels and mtDNA copy number in Pb and β-amyloid peptides (1-40) and (25-35) exposed cells. Also, significant reductions in the expression of mitochondrial electron transport chain (ETC) complex proteins (ATP5A, COXIV, UQCRC2, SDHB, NDUFS3), as well as down regulation of ETC complex gene expressions such as COXIV, ATP5F1 and NDUFS3 and antioxidant gene expressions like MnSOD and Gpx4 were observed in exposed cells. Furthermore, Pb and β-amyloid peptides exposure resulted in elevated mitochondrial malondialdehyde levels and a decrease in mitochondrial GSH levels. Our findings suggest that Pb toxicity could be one of the causative factors for the mitochondrial dysfunction and oxidative stress in Alzheimer's disease progression.
Collapse
Affiliation(s)
- Lakshmi Jaya Madhuri Bandaru
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Bindu Lasya C
- Department of Pharmacology, Anurag University, Hyderabad, India
| | | | - Suresh Challa
- Cell Biology Division, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India.
| |
Collapse
|
14
|
Nasiri L, Vaez-Mahdavi MR, Hassanpour H, Ghazanfari T, Kaboudanian Ardestani S, Askari N, Mohseni Majd MA, Rahimlou B. Increased serum lipofuscin associated with leukocyte telomere shortening in veterans: a possible role for sulfur mustard exposure in delayed-onset accelerated cellular senescence. Int Immunopharmacol 2023; 114:109549. [PMID: 36508921 DOI: 10.1016/j.intimp.2022.109549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Sulfur mustard (SM) is a toxic gas that causes chronic inflammation and oxidative stress leading to cell senescence. This study aimed to evaluate two indicators of biological aging (i.e., serum lipofuscin level and leukocyte telomere length) and assess their relationship based on the severity of SM exposure in the long term. METHODS The study was performed on two groups of male participants. 1) SM-exposed group (exposed to SM once in 1987), 73 volunteers. 2) Non-exposed group, 16 healthy volunteers. The SM-exposed group was categorized into three subgroups based on the severity of SM exposure and body damage (asymptom, mild, and severe). The blood sample was prepared from members of each group. The serum lipofuscin, TGF-β, malondialdehyde (MDA), c-reactive protein (CRP), and leukocyte telomere length (TL) were measured in all participants. RESULTS The MDA level was increased in the SM-exposed group (mean = 39.6 µM, SD = 16.5) compared to the non-exposed group (mean = 21.1 µM, SD = 10.3) (P < 0.05). The CRP level was also increased in the SM-exposed group (mean = 5.12 mg/l, SD = 3.36) compared to the non-exposed group (mean = 3.51 mg/l, SD = 1.21), while the TGF-β level was decreased (P < 0.05) in the SM-exposed group (mean = 52.6 pg/ml, SD = 18.7) compared to the non-exposed group (mean = 68.9 pg/ml, SD = 13.8). The relative TL was shorter in the SM-exposed group (mean = 0.40, SD = 0.28) than in the non-exposed group (mean = 2.25, SD = 1.41) (P < 0.05). The lipofuscin level was higher in the total SM-exposed group (mean = 1.44 ng/ml, SD = 0.685) than in the non-exposed group (mean = 0.88 ng/ml, SD = 0.449) (P < 0.05). The MDA and CRP levels were increased in the SM-exposed subgroups of asymptom, mild, and severe than the non-exposed group, while TGF-β level and TL were decreased in those subgroups. The lipofuscin level was higher in the SM-exposed subgroups of mild and severe than in the non-exposed group. The regression analysis determined a negative correlation between lipofuscin level and TL. The lipofuscin/TL ratio was higher in the total SM-exposed group (mean = 6.36, SD = 5.342) than in the non-exposed group (mean = 0.51, SD=0.389). This ratio was also higher in the SM-exposed subgroups of asymptom, mild, and severe than in the non-exposed group. The lipofuscin/TL ratio did not differ between mild and severe subgroups. CONCLUSION The delayed toxicity of SM is associated with chronic oxidative stress, continuous inflammatory stimulation, increased lipofuscin, and telomere shortening. Future studies are needed to verify the suitability of serum lipofuscin to telomere length ratio in determining the severity of SM toxicity.
Collapse
Affiliation(s)
- Leila Nasiri
- Department of Health Equity, Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Mohammad-Reza Vaez-Mahdavi
- Department of Health Equity, Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Physiology, Medical Faculty, Shahed University, Tehran, Iran.
| | - Hossein Hassanpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Sussan Kaboudanian Ardestani
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Nayere Askari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of biology, Faculty of Basic Sciences, Shahid Bahonar University, Kerman, Iran
| | | | - Bahman Rahimlou
- Department of Immunology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
15
|
Galper J, Kim WS, Dzamko N. LRRK2 and Lipid Pathways: Implications for Parkinson's Disease. Biomolecules 2022; 12:1597. [PMID: 36358947 PMCID: PMC9687231 DOI: 10.3390/biom12111597] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 04/10/2024] Open
Abstract
Genetic alterations in the LRRK2 gene, encoding leucine-rich repeat kinase 2, are a common risk factor for Parkinson's disease. How LRRK2 alterations lead to cell pathology is an area of ongoing investigation, however, multiple lines of evidence suggest a role for LRRK2 in lipid pathways. It is increasingly recognized that in addition to being energy reservoirs and structural entities, some lipids, including neural lipids, participate in signaling cascades. Early investigations revealed that LRRK2 localized to membranous and vesicular structures, suggesting an interaction of LRRK2 and lipids or lipid-associated proteins. LRRK2 substrates from the Rab GTPase family play a critical role in vesicle trafficking, lipid metabolism and lipid storage, all processes which rely on lipid dynamics. In addition, LRRK2 is associated with the phosphorylation and activity of enzymes that catabolize plasma membrane and lysosomal lipids. Furthermore, LRRK2 knockout studies have revealed that blood, brain and urine exhibit lipid level changes, including alterations to sterols, sphingolipids and phospholipids, respectively. In human LRRK2 mutation carriers, changes to sterols, sphingolipids, phospholipids, fatty acyls and glycerolipids are reported in multiple tissues. This review summarizes the evidence regarding associations between LRRK2 and lipids, and the functional consequences of LRRK2-associated lipid changes are discussed.
Collapse
Affiliation(s)
- Jasmin Galper
- Charles Perkins Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Woojin S Kim
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Nicolas Dzamko
- Charles Perkins Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
16
|
Hassan HM, Elnagar MR, Abdelrazik E, Mahdi MR, Hamza E, Elattar EM, ElNashar EM, Alghamdi MA, Al-Qahtani Z, Al-Khater KM, Aldahhan RA, ELdesoqui M. Neuroprotective effect of naringin against cerebellar changes in Alzheimer's disease through modulation of autophagy, oxidative stress and tau expression: An experimental study. Front Neuroanat 2022; 16:1012422. [PMID: 36312298 PMCID: PMC9615142 DOI: 10.3389/fnana.2022.1012422] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by gradual cognitive decline. Strong antioxidants that inhibit free radicals, such as polyphenols, reduce the likelihood of developing oxidative stress-related degenerative diseases such as AD. Naringin, a flavonoid found in citrus fruit shown to be neuroprotective, reduce oxidative damage and minimize histopathological changes caused by ischemic reperfusion, enhance the long-term memory in AD animal models. This work aimed to comprehend the role of naringin in the defense of the cerebellum against aluminum chloride (AlCl3)-induced AD in rats by investigating the behavioral, neurochemical, immunohistochemical, and molecular mechanisms that underpin its possible neuroprotective effects. Twenty-four adult albino rats were divided into four groups (n = 6/group): (i) Control (C) received saline per oral (p.o.), (ii) Naringin(N)-received naringin (100 mg/kg/d) p.o, (iii) AlCl3-recived AlCl3 (100 mg/kg/d) p.o and (iv) AlCl3 + Naringin (AlCl3 + N) received both AlCl3 and naringin p.o for 21 days. Behavioral tests showed an increase in the time to reach the platform in Morris water maze, indicating memory impairment in the AlCl3-treated group, but co-administration of naringin showed significant improvement. The Rotarod test demonstrated a decrease in muscle coordination in the AlCl3-treated group, while it was improved in the AlCl3 + N group. Neurochemical analysis of the hippocampus and cerebellum revealed that AlCl3 significantly increased lipid peroxidation and oxidative stress and decreased levels of reduced glutathione. Administration of naringin ameliorated these neurochemical changes via its antioxidant properties. Cerebellar immunohistochemical expression for microtubule assembly (tau protein) and oxidative stress (iNOS) increased in A1C13-treated group. On the other hand, the expression of the autophagic marker (LC3) in the cerebellum showed a marked decline in AlCl3-treated group. Western blot analysis confirmed the cerebellar immunohistochemical findings. Collectively, these findings suggested that naringin could contribute to the combat of oxidative and autophagic stress in the cerebellum of AlCl3-induced AD.
Collapse
Affiliation(s)
- Hend M. Hassan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed R. Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
- Department of Pharmacology, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Eman Abdelrazik
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed R. Mahdi
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Hamza
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Horus University, Damietta, Egypt
| | - Eman M. Elattar
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Eman Mohamed ElNashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha, Egypt
| | - Mansour Abdullah Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Zainah Al-Qahtani
- Neurology Section, Department of Internal Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Khulood Mohammed Al-Khater
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Rashid A. Aldahhan
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mamdouh ELdesoqui
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
K A, Mishra A, Singh S. Implications of intracellular protein degradation pathways in Parkinson's disease and therapeutics. J Neurosci Res 2022; 100:1834-1844. [PMID: 35819247 DOI: 10.1002/jnr.25101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/31/2022] [Accepted: 06/18/2022] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) pathology is the most common motor neurodegenerative disease that occurs due to the progressive degeneration of dopaminergic neurons of the nigrostriatal pathway of the brain. The histopathological hallmark of the disease is fibrillary aggregate called Lewy bodies which majorly contain α-synuclein, suggesting the critical implication of diminished protein degradation mechanisms in disease pathogenesis. This α-synuclein-containing Lewy bodies are evident in both experimental models as well as in postmortem PD brain and are speculated to be pathogenic but still, the lineal association between these aggregates and the complexity of disease pathology is not yet well established and needs further attention. However, it has been reported that α-synuclein aggregates have consorted with the declined proteasome and lysosome activities. Therefore, in this review, we reappraise intracellular protein degradation mechanisms during PD pathology. This article focused on the findings of the last two decades suggesting the implications of protein degradation mechanisms in disease pathogenesis and based on shreds of evidence, some of the approaches are also suggested which may be adopted to find out the novel therapeutic targets for the management of PD patients.
Collapse
Affiliation(s)
- Amrutha K
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
18
|
Simicic D, Cudalbu C, Pierzchala K. Overview of oxidative stress findings in hepatic encephalopathy: From cellular and ammonium-based animal models to human data. Anal Biochem 2022; 654:114795. [PMID: 35753389 DOI: 10.1016/j.ab.2022.114795] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
Oxidative stress is a natural phenomenon in the body. Under physiological conditions intracellular reactive oxygen species (ROS) are normal components of signal transduction cascades, and their levels are maintained by a complex antioxidants systems participating in the in-vivo redox homeostasis. Increased oxidative stress is present in several chronic diseases and interferes with phagocytic and nervous cell functions, causing an up-regulation of cytokines and inflammation. Hepatic encephalopathy (HE) occurs in both acute liver failure (ALF) and chronic liver disease. Increased blood and brain ammonium has been considered as an important factor in pathogenesis of HE and has been associated with inflammation, neurotoxicity, and oxidative stress. The relationship between ROS and the pathophysiology of HE is still poorly understood. Therefore, sensing ROS production for a better understanding of the relationship between oxidative stress and functional outcome in HE pathophysiology is critical for determining the disease mechanisms, as well as to improve the management of patients. This review is emphasizing the important role of oxidative stress in HE development and documents the changes occurring as a consequence of oxidative stress augmentation based on cellular and ammonium-based animal models to human data.
Collapse
Affiliation(s)
- D Simicic
- CIBM Center for Biomedical Imaging, Switzerland; Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Laboratory of Functional and Metabolic Imaging, EPFL, Lausanne, Switzerland
| | - C Cudalbu
- CIBM Center for Biomedical Imaging, Switzerland; Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - K Pierzchala
- CIBM Center for Biomedical Imaging, Switzerland; Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Laboratory of Functional and Metabolic Imaging, EPFL, Lausanne, Switzerland.
| |
Collapse
|
19
|
Xu J, Minobe E, Kameyama M. Ca2+ Dyshomeostasis Links Risk Factors to Neurodegeneration in Parkinson’s Disease. Front Cell Neurosci 2022; 16:867385. [PMID: 35496903 PMCID: PMC9050104 DOI: 10.3389/fncel.2022.867385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/23/2022] [Indexed: 12/06/2022] Open
Abstract
Parkinson’s disease (PD), a common neurodegenerative disease characterized by motor dysfunction, results from the death of dopaminergic neurons in the substantia nigra pars compacta (SNc). Although the precise causes of PD are still unknown, several risk factors for PD have been determined, including aging, genetic mutations, environmental factors, and gender. Currently, the molecular mechanisms underlying risk factor-related neurodegeneration in PD remain elusive. Endoplasmic reticulum stress, excessive reactive oxygen species production, and impaired autophagy have been implicated in neuronal death in the SNc in PD. Considering that these pathological processes are tightly associated with intracellular Ca2+, it is reasonable to hypothesize that dysregulation of Ca2+ handling may mediate risk factors-related PD pathogenesis. We review the recent findings on how risk factors cause Ca2+ dyshomeostasis and how aberrant Ca2+ handling triggers dopaminergic neurodegeneration in the SNc in PD, thus putting forward the possibility that manipulation of specific Ca2+ handling proteins and subcellular Ca2+ homeostasis may lead to new promising strategies for PD treatment.
Collapse
|
20
|
Stouffer MA, Khalaf-Nazzal R, Cifuentes-Diaz C, Albertini G, Bandet E, Grannec G, Lavilla V, Deleuze JF, Olaso R, Nosten-Bertrand M, Francis F. Doublecortin mutation leads to persistent defects in the Golgi apparatus and mitochondria in adult hippocampal pyramidal cells. Neurobiol Dis 2022; 168:105702. [PMID: 35339680 DOI: 10.1016/j.nbd.2022.105702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 11/08/2022] Open
Abstract
Human doublecortin (DCX) mutations are associated with severe brain malformations leading to aberrant neuron positioning (heterotopia), intellectual disability and epilepsy. DCX is a microtubule-associated protein which plays a key role during neurodevelopment in neuronal migration and differentiation. Dcx knockout (KO) mice show disorganized hippocampal pyramidal neurons. The CA2/CA3 pyramidal cell layer is present as two abnormal layers and disorganized CA3 KO pyramidal neurons are also more excitable than wild-type (WT) cells. To further identify abnormalities, we characterized Dcx KO hippocampal neurons at subcellular, molecular and ultrastructural levels. Severe defects were observed in mitochondria, affecting number and distribution. Also, the Golgi apparatus was visibly abnormal, increased in volume and abnormally organized. Transcriptome analyses from laser microdissected hippocampal tissue at postnatal day 60 (P60) highlighted organelle abnormalities. Ultrastructural studies of CA3 cells performed in P60 (young adult) and > 9 months (mature) tissue showed that organelle defects are persistent throughout life. Locomotor activity and fear memory of young and mature adults were also abnormal: Dcx KO mice consistently performed less well than WT littermates, with defects becoming more severe with age. Thus, we show that disruption of a neurodevelopmentally-regulated gene can lead to permanent organelle anomalies contributing to abnormal adult behavior.
Collapse
Affiliation(s)
- M A Stouffer
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - R Khalaf-Nazzal
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - C Cifuentes-Diaz
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - G Albertini
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - E Bandet
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - G Grannec
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - V Lavilla
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057 Evry, France
| | - J-F Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057 Evry, France
| | - R Olaso
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057 Evry, France
| | - M Nosten-Bertrand
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - F Francis
- INSERM UMR-S 1270, Paris 75005, France; Sorbonne Université, Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France.
| |
Collapse
|
21
|
Targeting autophagy, oxidative stress, and ER stress for neurodegenerative diseases treatment. J Control Release 2022; 345:147-175. [DOI: 10.1016/j.jconrel.2022.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/13/2022]
|
22
|
Biscaro RC, Mussi L, Sufi B, Padovani G, Camargo Junior FB, Magalhães WV, Di Stasi LC. Modulation of autophagy by an innovative phytocosmetic preparation (
Myrothamnus flabelifolia
and
Coffea arabica
) in human fibroblasts and its effects in a clinical randomized placebo‐controlled trial. J Cosmet Dermatol 2022; 21:4901-4912. [DOI: 10.1111/jocd.14888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/18/2022] [Accepted: 02/22/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Rafael C. Biscaro
- Research and Development Department Chemyunion Química Ltda Sorocaba Brazil
| | - Lilian Mussi
- Research and Development Department Chemyunion Química Ltda Sorocaba Brazil
| | - Bianca Sufi
- Research and Development Department Chemyunion Química Ltda Sorocaba Brazil
| | - Giovana Padovani
- Research and Development Department Chemyunion Química Ltda Sorocaba Brazil
| | | | | | - Luiz C. Di Stasi
- Laboratory of Phytomedicines, Pharmacology, and Biotechnology (PhytoPharmaTech) Department of Biophysics and Pharmacology Institute of Biosciences São Paulo State University (Unesp) Botucatu Brazil
| |
Collapse
|
23
|
Bandaru LJM, Ayyalasomayajula N, Murumulla L, Challa S. Mechanisms associated with the dysregulation of mitochondrial function due to lead exposure and possible implications on the development of Alzheimer's disease. Biometals 2022; 35:1-25. [PMID: 35048237 DOI: 10.1007/s10534-021-00360-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/09/2021] [Indexed: 01/17/2023]
Abstract
Lead (Pb) is a multimedia contaminant with various pathophysiological consequences, including cognitive decline and neural abnormalities. Recent findings have reported an association of Pb toxicity with Alzheimer's disease (AD). Studies have revealed that mitochondrial dysfunction is a pathological characteristic of AD. According to toxicology reports, Pb promotes mitochondrial oxidative stress by lowering complex III activity in the electron transport chain, boosting reactive oxygen species formation, and reducing the cell's antioxidant defence system. Here, we review recent advances in the role of mitochondria in Pb-induced AD pathology, as well as the mechanisms associated with the mitochondrial dysfunction, such as the depolarisation of the mitochondrial membrane potential, mitochondrial permeability transition pore opening; mitochondrial biogenesis, bioenergetics and mitochondrial dynamics alterations; and mitophagy and apoptosis. We also discuss possible therapeutic options for mitochondrial-targeted neurodegenerative disease (AD).
Collapse
Affiliation(s)
- Lakshmi Jaya Madhuri Bandaru
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Neelima Ayyalasomayajula
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Lokesh Murumulla
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India
| | - Suresh Challa
- Department of Cell Biology, National Institute of Nutrition, Indian Council of Medical Research (ICMR), Hyderabad, Telangana, 500007, India.
| |
Collapse
|
24
|
Ghozy S, Kacimi SEO, Elfil M, Sobeeh MG, Reda A, Kallmes KM, Rabinstein AA, Holmes DR, Brinjikji W, Kadirvel R, Kallmes DF. Transient Ischemic Attacks Preceding Ischemic Stroke and the Possible Preconditioning of the Human Brain: A Systematic Review and Meta-Analysis. Front Neurol 2021; 12:755167. [PMID: 34899573 PMCID: PMC8652229 DOI: 10.3389/fneur.2021.755167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
Stroke is a leading cause of mortality and disability worldwide. Transient ischemic attack (TIA) is defined as transient brain ischemia with temporary neurological deficits. In animal models, prior TIA seems to enhance brain ischemic tolerance to withstand further ischemic events, which might be explained by brain preconditioning. Thus, this review aims to formulate evidence of whether TIAs can induce positive preconditioning and enhance the functional outcomes in patients suffering from subsequent ischemic strokes. Five databases were searched (PubMed, Embase, SAGE, Web of Science, and Scopus), and twelve studies were included in the quantitative analysis. Studies were eligible when comparing patients with acute ischemic stroke (AIS) and previous TIA with those with AIS without TIA. Comparisons included the National Institute of Health Stroke Scale (NIHSS) score at admission and 7 days from the stroke event, modified Rankin score (mRS), and Trial of ORG 10,172 in Acute Stroke Treatment (TOAST) classification. Odds ratio (OR), mean difference (MD), and 95% confidence interval (CI) were used to describe our results using the random effect model. Our results revealed that patients with stroke and prior TIAs had lower NIHSS scores at admission than those without prior TIAs. However, the NIHSS score was not significantly different between the two groups at 7 days. Furthermore, there was no statistically significant difference between both groups in terms of mortality. Despite the differences in the admission mRS score groups, patients with prior TIAs had lower mRS scores at discharge.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | | | - Mohammed Elfil
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Cairo University, Cairo, Egypt.,Faculty of Physical Therapy, Sinai University, Cairo, Egypt
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Kevin M Kallmes
- Nested Knowledge, St. Paul, MN, United States.,Superior Medical Experts, St. Paul, MN, United States
| | - Alejandro A Rabinstein
- Department of Neurology and Neurocritical Care, Mayo Clinic, Rochester, MN, United States
| | - David R Holmes
- Department of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Waleed Brinjikji
- Department of Radiology, Mayo Clinic, Rochester, MN, United States.,Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
25
|
Parella KJ, Manhardt C, Capucilli D, Moyer B, Colegrove H, Moody KJ, Sleeper M, Banas A, Rebbaa A, Wolfe AJ. Fluorescence-Based Detection of Ferrous Iron in Senescent Cells. Rejuvenation Res 2021; 24:456-463. [PMID: 34841899 DOI: 10.1089/rej.2021.0075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A major limitation in aging research is the lack of reliable biomarkers to assess phenotypic changes with age or monitor response to antiaging interventions. This study investigates the role of intracellular ferrous iron (Fe2+) as a potential biomarker of senescence. Iron is known to accumulate in various tissues with age and recent studies have demonstrated that its level increases dramatically in senescent cells. The current techniques used to measure the accumulation of iron are cumbersome and only measure total iron not specific isotopes such as the redox reactive Fe2+. It is still to be determined whether the damaging form of iron (Fe2+) is specifically elevated in senescent cells. In this study, we assessed the potential use of a newly discovered Fe2+ reactive probe (SiRhoNox-1) for selective labeling of senescent cells in vitro. For this we have generated various senescent cell models and subjected them to SiRhoNox-1 labeling. Our results indicate that SiRhoNox-1 selectivity labels live senescent cells and was more specific and faster than current staining such as SA-βGal or a derived fluorescent probe C12FDG. Together these findings suggest that SiRhoNox-1 may serve as a convenient tool to detect senescent cells based on their ferrous iron level.
Collapse
Affiliation(s)
- Kyle J Parella
- Ichor Life Sciences, Inc., Lafayette, New York, USA.,Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA.,Department of Chemistry, Clarkson University, Potsdam, New York, USA
| | | | | | | | | | - Kelsey J Moody
- Ichor Life Sciences, Inc., Lafayette, New York, USA.,Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA.,Department of Chemistry, Clarkson University, Potsdam, New York, USA.,FoxBio, Inc., Lafayette, New York, USA
| | | | - Andrew Banas
- Ichor Life Sciences, Inc., Lafayette, New York, USA
| | | | - Aaron J Wolfe
- Ichor Life Sciences, Inc., Lafayette, New York, USA.,Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA.,Department of Chemistry, Clarkson University, Potsdam, New York, USA.,FoxBio, Inc., Lafayette, New York, USA
| |
Collapse
|
26
|
The Glyoxalase System in Age-Related Diseases: Nutritional Intervention as Anti-Ageing Strategy. Cells 2021; 10:cells10081852. [PMID: 34440621 PMCID: PMC8393707 DOI: 10.3390/cells10081852] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/19/2022] Open
Abstract
The glyoxalase system is critical for the detoxification of advanced glycation end-products (AGEs). AGEs are toxic compounds resulting from the non-enzymatic modification of biomolecules by sugars or their metabolites through a process called glycation. AGEs have adverse effects on many tissues, playing a pathogenic role in the progression of molecular and cellular aging. Due to the age-related decline in different anti-AGE mechanisms, including detoxifying mechanisms and proteolytic capacities, glycated biomolecules are accumulated during normal aging in our body in a tissue-dependent manner. Viewed in this way, anti-AGE detoxifying systems are proposed as therapeutic targets to fight pathological dysfunction associated with AGE accumulation and cytotoxicity. Here, we summarize the current state of knowledge related to the protective mechanisms against glycative stress, with a special emphasis on the glyoxalase system as the primary mechanism for detoxifying the reactive intermediates of glycation. This review focuses on glyoxalase 1 (GLO1), the first enzyme of the glyoxalase system, and the rate-limiting enzyme of this catalytic process. Although GLO1 is ubiquitously expressed, protein levels and activities are regulated in a tissue-dependent manner. We provide a comparative analysis of GLO1 protein in different tissues. Our findings indicate a role for the glyoxalase system in homeostasis in the eye retina, a highly oxygenated tissue with rapid protein turnover. We also describe modulation of the glyoxalase system as a therapeutic target to delay the development of age-related diseases and summarize the literature that describes the current knowledge about nutritional compounds with properties to modulate the glyoxalase system.
Collapse
|
27
|
Liu Y, Hu J, Yao S, Zhou Q, Li H, Takahata T. Multiple Visuotopically Organized Subdivisions of the Lateral Pulvinar/Central Lateral Inferior Pulvinar Project into Thin and Thick Stripe Compartments of V2 in Macaques. Cereb Cortex 2021; 31:3788-3803. [PMID: 33772553 DOI: 10.1093/cercor/bhab049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/30/2021] [Accepted: 02/13/2021] [Indexed: 11/13/2022] Open
Abstract
The lateral and central lateral inferior pulvinar (PL/PIcl) of primates has been implicated in playing an important role in visual processing, but its physiological and anatomical characteristics remain to be elucidated. It has been suggested that there are two complete visuotopic maps in the PL/PIcl, each of which sends afferents into V2 and V4 in primates. Given that functionally distinct thin and thick stripes of V2 both receive inputs from the PL/PIcl, this raises the possibility of a presence of parallel segregated pathways within the PL/PIcl. To address this question, we selectively injected three types of retrograde tracers (CTB-488, CTB-555, and BDA) into thin or thick stripes in V2 and examined labeling in the PL/PIcl in macaques. As a result, we found that every cluster of retrograde labeling in the PL/PIcl included all three types of signals next to each other, suggesting that thin stripe- and thick stripe-projecting compartments are not segregated into domains. Unexpectedly, we found at least five topographically organized retrograde labeling clusters in the PL/PIcl, indicating the presence of more than two V2-projecting maps. Our results suggest that the PL/PIcl exhibits greater compartmentalization than previously thought. They may be functionally similar but participate in multiple cortico-pulvinar-cortical loops.
Collapse
Affiliation(s)
- Ye Liu
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, P. R. China
| | - Jiaming Hu
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, P. R. China
| | - Songping Yao
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, P. R. China.,Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University,Hangzhou 310029, P. R. China
| | - Qiuying Zhou
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, P. R. China
| | - Hangqi Li
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, P. R. China.,Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University,Hangzhou 310029, P. R. China
| | - Toru Takahata
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou 310029, P. R. China.,Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University,Hangzhou 310029, P. R. China
| |
Collapse
|
28
|
TFEB protein expression is reduced in aged brains and its overexpression mitigates senescence-associated biomarkers and memory deficits in mice. Neurobiol Aging 2021; 106:26-36. [PMID: 34229273 DOI: 10.1016/j.neurobiolaging.2021.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 05/18/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022]
Abstract
Identification of molecules and molecular pathways that can ameliorate aging-associated decline in cognitive function is crucial. Here we report that the protein levels of transcription factor EB (TFEB) were markedly reduced in both the cytosolic and nuclear fractions of the frontal cortex and hippocampus at 18-months of age relative to 6 months in the normal male wild-type mice. In the transgenic mice with ectopic expression of flag-TFEB in neurons, we observed that the levels of actin-normalized PGC1α and mtTFA were significantly increased in both the cortex and the hippocampus. Additionally, we confirmed increased mitochondria numbers in the flag-TFEB mice by transmission electron microscopy. Most importantly, TFEB expression in the 18-month-old transgenic mice mitigated markers of senescence including P16INK4a, γ-H2AX, and lamin B1, and improved memory skills implying that TFEB may exert an anti-aging effect by modulating neuronal senescence. Taken together these data strongly support that TFEB can be a useful therapeutic target for brain senescent cells to help overcome the age-related issues in cognition and possibly, achieve healthy aging.
Collapse
|
29
|
Perluigi M, Di Domenico F, Barone E, Butterfield DA. mTOR in Alzheimer disease and its earlier stages: Links to oxidative damage in the progression of this dementing disorder. Free Radic Biol Med 2021; 169:382-396. [PMID: 33933601 PMCID: PMC8145782 DOI: 10.1016/j.freeradbiomed.2021.04.025] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly population and has worldwide impact. The etiology of the disease is complex and results from the confluence of multiple mechanisms ultimately leading to neuronal loss and cognitive decline. Among risk factors, aging is the most relevant and accounts for several pathogenic events that contribute to disease-specific toxic mechanisms. Accumulating evidence linked the alterations of the mammalian target of rapamycin (mTOR), a serine/threonine protein kinase playing a key role in the regulation of protein synthesis and degradation, to age-dependent cognitive decline and pathogenesis of AD. To date, growing studies demonstrated that aberrant mTOR signaling in the brain affects several pathways involved in energy metabolism, cell growth, mitochondrial function and proteostasis. Recent advances associated alterations of the mTOR pathway with the increased oxidative stress. Disruption of all these events strongly contribute to age-related cognitive decline including AD. The current review discusses the main regulatory roles of mTOR signaling network in the brain, focusing on its role in autophagy, oxidative stress and energy metabolism. Collectively, experimental data suggest that targeting mTOR in the CNS can be a valuable strategy to prevent/slow the progression of AD.
Collapse
Affiliation(s)
- M Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - F Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - E Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D A Butterfield
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy; Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
30
|
Cao W, Li J, Yang K, Cao D. An overview of autophagy: Mechanism, regulation and research progress. Bull Cancer 2021; 108:304-322. [PMID: 33423775 DOI: 10.1016/j.bulcan.2020.11.004] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Autophagy refers to the formation of autophagosomes by membrane wrapping part of the cytoplasm and the organelles and proteins that need to be degraded in the cells. Autophagosomes are fused with lysosomes to form autophagolysosome, which degrade the contents of the inclusions, to achieve cell homeostasis and organelle renewal. The regulatory mechanism of autophagy is complex, and its upstream signaling pathway mainly involves mTOR dependent pathway and mTOR independent pathway (AMPK, PI3K, Ras-MAPK, p53, PTEN, endoplasmic reticulum stress). Autophagy is a phenomenon of "self-eating" in cells. Apoptosis is a phenomenon of "self-killing". Both of them share the same stimulating factors and regulatory proteins, but the threshold of induction is different. How to transform and coordinate is not clear at present. This paper summarizes the history of autophagy discovery, the structure and function of related molecules, the biological function of autophagy, the regulatory mechanism and the research results of the relationship between autophagy and apoptosis.
Collapse
Affiliation(s)
- Weiya Cao
- Anhui University of Science & Technology, Medical school, Huainan 232001, China.
| | - Jinhong Li
- Juancheng Hospital of Shandong Provincial Hospital Group, Heze 274100, China
| | - Kepeng Yang
- Anhui University of Science & Technology, Medical school, Huainan 232001, China
| | - Dongli Cao
- Anhui University of Science & Technology, Medical school, Huainan 232001, China
| |
Collapse
|
31
|
Finkbeiner S. Functional genomics, genetic risk profiling and cell phenotypes in neurodegenerative disease. Neurobiol Dis 2020; 146:105088. [PMID: 32977020 PMCID: PMC7686089 DOI: 10.1016/j.nbd.2020.105088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/03/2022] Open
Abstract
Human genetics provides unbiased insights into the causes of human disease, which can be used to create a foundation for effective ways to more accurately diagnose patients, stratify patients for more successful clinical trials, discover and develop new therapies, and ultimately help patients choose the safest and most promising therapeutic option based on their risk profile. But the process for translating basic observations from human genetics studies into pathogenic disease mechanisms and treatments is laborious and complex, and this challenge has particularly slowed the development of interventions for neurodegenerative disease. In this review, we discuss the many steps in the process, the important considerations at each stage, and some of the latest tools and technologies that are available to help investigators translate insights from human genetics into diagnostic and therapeutic strategies that will lead to the sort of advances in clinical care that make a difference for patients.
Collapse
Affiliation(s)
- Steven Finkbeiner
- Center for Systems and Therapeutics, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Neurology and Physiology, University of Califorina, San Francisco, CA 94158, USA.
| |
Collapse
|
32
|
Moore MN. Lysosomes, Autophagy, and Hormesis in Cell Physiology, Pathology, and Age-Related Disease. Dose Response 2020; 18:1559325820934227. [PMID: 32684871 PMCID: PMC7343375 DOI: 10.1177/1559325820934227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/02/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
Autophagy has been strongly linked with hormesis, however, it is only relatively recently that the mechanistic basis underlying this association has begun to emerge. Lysosomal autophagy is a group of processes that degrade proteins, protein aggregates, membranes, organelles, segregated regions of cytoplasm, and even parts of the nucleus in eukaryotic cells. These degradative processes are evolutionarily very ancient and provide a survival capability for cells that are stressed or injured. Autophagy and autophagic dysfunction have been linked with many aspects of cell physiology and pathology in disease processes; and there is now intense interest in identifying various therapeutic strategies involving its regulation. The main regulatory pathway for augmented autophagy is the mechanistic target of rapamycin (mTOR) cell signaling, although other pathways can be involved, such as 5'-adenosine monophosphate-activated protein kinase. Mechanistic target of rapamycin is a key player in the many highly interconnected intracellular signaling pathways and is responsible for the control of cell growth among other processes. Inhibition of mTOR (specifically dephosphorylation of mTOR complex 1) triggers augmented autophagy and the search is on the find inhibitors that can induce hormetic responses that may be suitable for treating many diseases, including many cancers, type 2 diabetes, and age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Michael N. Moore
- European Centre for Environment & Human Health (ECEHH), University of Exeter Medical School, Knowledge Spa, Royal Cornwall Hospital, Truro, United Kingdom
- Plymouth Marine Laboratory, Plymouth, Devon, United Kingdom
- School of Biological & Marine Sciences, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
33
|
Dhakal S, Macreadie I. Tyramine and Amyloid Beta 42: A Toxic Synergy. Biomedicines 2020; 8:biomedicines8060145. [PMID: 32486277 PMCID: PMC7345151 DOI: 10.3390/biomedicines8060145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022] Open
Abstract
Implicated in various diseases including Parkinson's disease, Huntington's disease, migraines, schizophrenia and increased blood pressure, tyramine plays a crucial role as a neurotransmitter in the synaptic cleft by reducing serotonergic and dopaminergic signaling through a trace amine-associated receptor (TAAR1). There appear to be no studies investigating a connection of tyramine to Alzheimer's disease. This study aimed to examine whether tyramine could be involved in AD pathology by using Saccharomyces cerevisiae expressing Aβ42. S. cerevisiae cells producing native Aβ42 were treated with different concentrations of tyramine, and the production of reactive oxygen species (ROS) was evaluated using flow cytometric cell analysis. There was dose-dependent ROS generation in wild-type yeast cells with tyramine. In yeast producing Aβ42, ROS levels generated were significantly higher than in controls, suggesting a synergistic toxicity of Aβ42 and tyramine. The addition of exogenous reduced glutathione (GSH) was found to rescue the cells with increased ROS, indicating depletion of intracellular GSH due to tyramine and Aβ42. Additionally, tyramine inhibited the respiratory growth of yeast cells producing GFP-Aβ42, while there was no growth inhibition when cells were producing GFP. Tyramine was also demonstrated to cause increased mitochondrial DNA damage, resulting in the formation of petite mutants that lack respiratory function. These findings indicate that there can be a detrimental synergy between Aβ42 and tyramine, which could be considered in Alzheimer's disease. This work also demonstrates the utility of yeast as a model for studying toxic agents such as Aβ42, tyramine, and agents that might exacerbate AD pathology.
Collapse
|
34
|
Moore MN, Shaw JP, Pascoe C, Beesley A, Viarengo A, Lowe DM. Anti-oxidative hormetic effects of cellular autophagy induced by nutrient deprivation in a molluscan animal model. MARINE ENVIRONMENTAL RESEARCH 2020; 156:104903. [PMID: 32056801 DOI: 10.1016/j.marenvres.2020.104903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/24/2020] [Accepted: 02/03/2020] [Indexed: 06/10/2023]
Abstract
This investigation using a molluscan animal model tested the hypothesis that experimentally induced lysosomal autophagy protects against oxidative cell injury. Induction of augmented lysosomal autophagy has previously been implicated in this protective process. Four treatment groups of blue mussels (Mytilus galloprovincialis) were used: Group 1 (fed - control), Group 2 (fasted), Group 3 (copper + fed) and Group 4 (copper + fasted). Groups 2 and 4 were fasted in order to trigger autophagy; and samples of hepatopancreas (liver analogue or digestive gland) from all 4 groups were taken at 3, 6 and 15 days. Treatment with copper provided a positive reference for oxidative stress: Groups 3 and 4 were treated with copper (10 μg Cu2+/animal/day) for three days only. Oxidative damage and cellular injury in hepatopancreatic digestive cells was found to decrease in Group 2 (fasted) compared to Group 1 (fed - control). Group 3 (fed + copper) showed clear evidence of oxidative stress and cell injury, as well as induction of antioxidant activities. Group 4 (copper + fasted) had a reduced uptake of copper and toxicity of copper was also reduced, compared with Group 3. It was concluded that augmented autophagy had a hormetic cytoprotective anti-oxidant effect.
Collapse
Affiliation(s)
- M N Moore
- Plymouth Marine Laboratory, Prospect Place, The Hoe, Plymouth, Devon, PL1 3DH, UK; European Centre for Environment & Human Health (ECEHH), University of Exeter Medical School, Knowledge Spa, Royal Cornwall Hospital, Truro, TR1 3HD, UK; School of Biological & Marine Sciences, University of Plymouth, Drake Circus, Plymouth, PL4 8AA, UK.
| | - J P Shaw
- Plymouth Marine Laboratory, Prospect Place, The Hoe, Plymouth, Devon, PL1 3DH, UK
| | - C Pascoe
- Plymouth Marine Laboratory, Prospect Place, The Hoe, Plymouth, Devon, PL1 3DH, UK
| | - A Beesley
- Plymouth Marine Laboratory, Prospect Place, The Hoe, Plymouth, Devon, PL1 3DH, UK
| | - A Viarengo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via La Masa 19-20156, Milano, Italy
| | - D M Lowe
- Plymouth Marine Laboratory, Prospect Place, The Hoe, Plymouth, Devon, PL1 3DH, UK
| |
Collapse
|
35
|
Finkbeiner S. The Autophagy Lysosomal Pathway and Neurodegeneration. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a033993. [PMID: 30936119 DOI: 10.1101/cshperspect.a033993] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The autophagy lysosomal pathway (ALP) is a major mechanism for degrading intracellular macromolecules. The catabolic products can then be used by the cell for energy or as building blocks to make other macromolecules. Since its discovery, a variety of cellular pathways have emerged that target components with varying specificity for lysosomal degradation. Under some circumstances, lysosomes may release their contents into the extracellular space where they may serve signaling or pathogenic functions. The ALP is active in healthy cells, and the level of activity can be regulated by nutrient-sensing and metabolic signaling pathways. The ALP is the primary pathway by which lipids and damaged organelles are degraded and may be the only pathway capable of degrading aggregated proteins. As such, there has been intense interest in understanding the role of the ALP in the accumulation of aggregated misfolded proteins characteristic of many of the major adult-onset neurodegenerative diseases. This review focuses on recent advances in our understanding of the ALP and its potential relationship to the pathogenesis and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Steven Finkbeiner
- Gladstone Institutes, San Francisco, California 94158.,Departments of Neurology and Physiology, University of California, San Francisco, California 94158
| |
Collapse
|
36
|
Joshi V, Upadhyay A, Prajapati VK, Mishra A. How autophagy can restore proteostasis defects in multiple diseases? Med Res Rev 2020; 40:1385-1439. [PMID: 32043639 DOI: 10.1002/med.21662] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/03/2020] [Accepted: 01/28/2020] [Indexed: 12/12/2022]
Abstract
Cellular evolution develops several conserved mechanisms by which cells can tolerate various difficult conditions and overall maintain homeostasis. Autophagy is a well-developed and evolutionarily conserved mechanism of catabolism, which endorses the degradation of foreign and endogenous materials via autolysosome. To decrease the burden of the ubiquitin-proteasome system (UPS), autophagy also promotes the selective degradation of proteins in a tightly regulated way to improve the physiological balance of cellular proteostasis that may get perturbed due to the accumulation of misfolded proteins. However, the diverse as well as selective clearance of unwanted materials and regulations of several cellular mechanisms via autophagy is still a critical mystery. Also, the failure of autophagy causes an increase in the accumulation of harmful protein aggregates that may lead to neurodegeneration. Therefore, it is necessary to address this multifactorial threat for in-depth research and develop more effective therapeutic strategies against lethal autophagy alterations. In this paper, we discuss the most relevant and recent reports on autophagy modulations and their impact on neurodegeneration and other complex disorders. We have summarized various pharmacological findings linked with the induction and suppression of autophagy mechanism and their promising preclinical and clinical applications to provide therapeutic solutions against neurodegeneration. The conclusion, key questions, and future prospectives sections summarize fundamental challenges and their possible feasible solutions linked with autophagy mechanism to potentially design an impactful therapeutic niche to treat neurodegenerative diseases and imperfect aging.
Collapse
Affiliation(s)
- Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Karwar, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Karwar, India
| | - Vijay K Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Karwar, India
| |
Collapse
|
37
|
Zhang C, Huo S, Fan Y, Gao Y, Yang Y, Sun D. Autophagy May Be Involved in Fluoride-Induced Learning Impairment in Rats. Biol Trace Elem Res 2020; 193:502-507. [PMID: 31111310 DOI: 10.1007/s12011-019-01735-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/22/2019] [Indexed: 12/30/2022]
Abstract
Fluoride can induce neurotoxicity, but the mechanism is not clear. In this study, we explored the role of autophagy in F--induced neurotoxicity of Wistar rats. Eighty Wistar rats were randomly divided into four groups: the control group (distilled water containing less than 0.1 mg/L F-) and three NaF-treated groups (F- was respectively administered at 25, 50, and 100 mg/L orally via drinking water). The water maze experiment showed that NaF exposure impaired the learning capabilities of the rats. When compared with the control group, the mean escape latency of the rats in the 100 mg/L F- group was much longer (P < 0.05). Immunohistochemical analysis showed that NaF exposure induced autophagy, as shown by the significant increase of Beclin-1 expression in the hippocampal CA1 region and DG region. Transmission electron microscopy was used to observe the ultrastructural changes of hippocampal neurons. With the increase of F- concentration, the ultrastructural abnormalities of hippocampal neurons increased. These results indicate that fluoride can impair the learning ability of rats, which may be related to the induction of autophagy in rat hippocampal neurons.
Collapse
Affiliation(s)
- Chengzhi Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin, 150081, China
| | - Simeng Huo
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin, 150081, China
| | - Yumei Fan
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin, 150081, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin, 150081, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin, 150081, China
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin, 150081, China.
| |
Collapse
|
38
|
Murillo AG, Hu S, Fernandez ML. Zeaxanthin: Metabolism, Properties, and Antioxidant Protection of Eyes, Heart, Liver, and Skin. Antioxidants (Basel) 2019; 8:E390. [PMID: 31514298 PMCID: PMC6770730 DOI: 10.3390/antiox8090390] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/31/2022] Open
Abstract
Zeaxanthin, a non-provitamin A carotenoid that belongs to the xanthophyll family, has been less studied than its isomer lutein. However, zeaxanthin has also been shown to have a number of beneficial effects for human health due to its ability to quench free radicals, exert antioxidant effects, as well as decrease inflammation. It is the purpose of this review to discuss the metabolism of zeaxanthin, including digestion, absorption, transport, and uptake by tissues, as well as the dietary or other factors which affect zeaxanthin bioavailability. In addition, this review also focuses on specific effects of this carotenoid on eye, skin, liver, and cardiovascular health. Data derived from human interventions, animal models of research, and in vitro and cell studies are discussed in this review.
Collapse
Affiliation(s)
- Ana Gabriela Murillo
- Biochemistry Department, University of Costa Rica, San Pedro de Montes de Oca 2060, Costa Rica.
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | - Siqi Hu
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
39
|
Khalifeh M, Barreto GE, Sahebkar A. Trehalose as a promising therapeutic candidate for the treatment of Parkinson's disease. Br J Pharmacol 2019; 176:1173-1189. [PMID: 30767205 PMCID: PMC6468260 DOI: 10.1111/bph.14623] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a progressive movement disorder resulting primarily from loss of nigrostriatal dopaminergic neurons. PD is characterized by the accumulation of protein aggregates, and evidence suggests that aberrant protein deposition in dopaminergic neurons could be related to the dysregulation of the lysosomal autophagy pathway. The therapeutic potential of autophagy modulators has been reported in experimental models of PD. Trehalose is a natural disaccharide that has been considered as a new candidate for the treatment of neurodegenerative diseases. It has a chaperone-like activity, prevents protein misfolding or aggregation, and by promoting autophagy, contributes to the removal of accumulated proteins. In this review, we briefly summarize the role of aberrant autophagy in PD and the underlying mechanisms that lead to the development of this disease. We also discuss reports that used trehalose to counteract the neurotoxicity in PD, focusing particularly on the autophagy promoting, protein stabilization, and anti-neuroinflammatory effects of trehalose.
Collapse
Affiliation(s)
- Masoomeh Khalifeh
- Department of Medical Biotechnology, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - George E. Barreto
- Departamento de Nutrición y Bioquímica, Facultad de CienciasPontificia Universidad JaverianaBogotáColombia
- Instituto de Ciencias BiomédicasUniversidad Autónoma de ChileSantiagoChile
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research CenterMashhad University of Medical SciencesMashhadIran
- Biotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- School of PharmacyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
40
|
Ding Q, Zhu H. Upregulation of PSMB8 and cathepsins in the human brains of dementia with Lewy bodies. Neurosci Lett 2019; 678:131-137. [PMID: 29775672 DOI: 10.1016/j.neulet.2018.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 01/08/2023]
Abstract
Proteasome and lysosome are responsible for the homeostasis of proteins, lipids and carbohydrates in cells. Numerous reports indicate the proteolytic pathways have altered functions during neurodegeneration and aging. Dementia with Lewy bodies (DLB) is one of the leading forms of dementia, and the proteolytic alteration in DLB has not yet been fully investigated. This study shows that the components of proteasome and lysosome had selectively altered gene expression and enzymatic functions. Specifically, PSMB8, an inducible proteasomal β subunit, had elevated mRNA level and protein level in DLB brain compared with age-matched controls. The proteasomal caspase-like peptidase showed significant decreased activity in DLB brains and the trypsin-like/chemotrypsin-like activities did not reach statistical significance. Lysosomal cathepsin B and D had elevated mRNA levels while only cathepsin B showed elevated enzymatic activity in DLB brains. This data indicate that the alteration of proteolytic pathways is highly selective and comprehensive. Further study to elucidate the correlation between neurodegenerative development and the alteration of proteolytic pathways would be important for therapeutic development.
Collapse
Affiliation(s)
- Qunxing Ding
- Department of Biological Sciences, Kent State University at East Liverpool, East Liverpool, OH 43920, USA.
| | - Haiyan Zhu
- Department of Biological Sciences, Kent State University at East Liverpool, East Liverpool, OH 43920, USA
| |
Collapse
|
41
|
Zhao Y, Zhang W, Jia Q, Feng Z, Guo J, Han X, Liu Y, Shang H, Wang Y, Liu WJ. High Dose Vitamin E Attenuates Diabetic Nephropathy via Alleviation of Autophagic Stress. Front Physiol 2019; 9:1939. [PMID: 30719008 PMCID: PMC6348272 DOI: 10.3389/fphys.2018.01939] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
It has been reported that autophagic stress, which is involved in many diseases, plays a key role in the development of diabetic nephropathy (DN). In this study, we investigated the effects of high dose vitamin E on renal tubular epithelial cells and autophagic stress-related mechanisms in diabetes condition. In diabetic rats, high dose vitamin E treatment significantly decreased the serum creatinine, urea nitrogen, urinary albumin and urinary protein, reduced the levels of LCN2, HAVCR1, LDH and 8-OHdG in urine, and attenuated the cellular apoptosis and interstitial fibrosis in renal cortex. In vitro, vitamin E could reduce the release of LCN2 and HAVCR1 and the protein levels of caspase 3 and TGF-β1, as well as improve the growth inhibition in cultured HK-2 cells after exposure to advanced glycation end products (AGEs). Also, LC3-II and SQSTM1-positive dots were significantly increased in the renal tubular epithelial cells of DN patients and diabetic rats, and in HK-2 cells after exposure to AGEs, which were markedly declined by vitamin E. In addition, we found that the autophagosome formation was not affected by AGEs, as assessed by the mRNA levels of LC3B, Beclin-1, and ATG7. However, AGEs blocked the lysosomal degradation of autophagosome, which was characterized by a decrease in the enzymatic activity of cathepsin B/cathepsin L and DQ-ovalbumin degradation in HK-2 cells, indicating that AGEs-induced accumulation of autophagic vacuoles was a sign of autophagic stress. Interestingly, vitamin E exerted a protective effect on lysosomes to reduce the autophagic stress. Taken together, we conclude that autophagic stress may play an important part in the progression of DN, and alleviation of autophagic stress though improvement of lysosomal function provides a promising novel approach for treating DN.
Collapse
Affiliation(s)
- Yuxue Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wenting Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Qi Jia
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Zhendong Feng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xueting Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yuning Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yaoxian Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.,Renal Research Institution of Beijing University of Chinese Medicine, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
42
|
Jové M, Pradas I, Dominguez-Gonzalez M, Ferrer I, Pamplona R. Lipids and lipoxidation in human brain aging. Mitochondrial ATP-synthase as a key lipoxidation target. Redox Biol 2018; 23:101082. [PMID: 30635167 PMCID: PMC6859548 DOI: 10.1016/j.redox.2018.101082] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/04/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022] Open
Abstract
The human brain is a target of the aging process like other cell systems of the human body. Specific regions of the human brain exhibit differential vulnerabilities to the aging process. Yet the underlying mechanisms that sustain the preservation or deterioration of neurons and cerebral functions are unknown. In this review, we focus attention on the role of lipids and the importance of the cross-regionally different vulnerabilities in human brain aging. In particular, we first consider a brief approach to the lipidomics of human brain, the relationship between lipids and lipoxidative damage, the role of lipids in human brain aging, and the specific targets of lipoxidative damage in human brain and during aging. It is proposed that the restricted set of modified proteins and the functional categories involved may be considered putative collaborative factors contributing to neuronal aging, and that mitochondrial ATP synthase is a key lipoxidative target in human brain aging.
Collapse
Affiliation(s)
- Mariona Jové
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Irene Pradas
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain
| | - Mayelin Dominguez-Gonzalez
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona; Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Institute for Research in Biomedicine of Lleida (UdL-IRBLleida), Lleida, Spain.
| |
Collapse
|
43
|
Fimognari N, Hollings A, Lam V, Tidy RJ, Kewish CM, Albrecht MA, Takechi R, Mamo JCL, Hackett MJ. Biospectroscopic Imaging Provides Evidence of Hippocampal Zn Deficiency and Decreased Lipid Unsaturation in an Accelerated Aging Mouse Model. ACS Chem Neurosci 2018; 9:2774-2785. [PMID: 29901988 DOI: 10.1021/acschemneuro.8b00193] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Western society is facing a health epidemic due to the increasing incidence of dementia in aging populations, and there are still few effective diagnostic methods, minimal treatment options, and no cure. Aging is the greatest risk factor for memory loss that occurs during the natural aging process, as well as being the greatest risk factor for neurodegenerative disease such as Alzheimer's disease. Greater understanding of the biochemical pathways that drive a healthy aging brain toward dementia (pathological aging or Alzheimer's disease), is required to accelerate the development of improved diagnostics and therapies. Unfortunately, many animal models of dementia model chronic amyloid precursor protein overexpression, which although highly relevant to mechanisms of amyloidosis and familial Alzheimer's disease, does not model well dementia during the natural aging process. A promising animal model reported to model mechanisms of accelerated natural aging and memory impairments, is the senescence accelerated murine prone strain 8 (SAMP8), which has been adopted by many research group to study the biochemical transitions that occur during brain aging. A limitation to traditional methods of biochemical characterization is that many important biochemical and elemental markers (lipid saturation, lactate, transition metals) cannot be imaged at meso- or microspatial resolution. Therefore, in this investigation, we report the first multimodal biospectroscopic characterization of the SAMP8 model, and have identified important biochemical and elemental alterations, and colocalizations, between 4 month old SAMP8 mice and the relevant control (SAMR1) mice. Specifically, we demonstrate direct evidence of Zn deficiency within specific subregions of the hippocampal CA3 sector, which colocalize with decreased lipid unsaturation. Our findings also revealed colocalization of decreased lipid unsaturation and increased lactate in the corpus callosum white matter, adjacent to the hippocampus. Such findings may have important implication for future research aimed at elucidating specific biochemical pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Nicholas Fimognari
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Biomedical Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Ashley Hollings
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Science, Curtin University, Bentley, WA 6845, Australia
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Public Health, Curtin University, Bentley, WA 6102, Australia
| | - Rebecca J. Tidy
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Science, Curtin University, Bentley, WA 6845, Australia
| | - Cameron M. Kewish
- Australian Nuclear Science and Technology Organisation, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Matthew A. Albrecht
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Ryu Takechi
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Public Health, Curtin University, Bentley, WA 6102, Australia
| | - John C. L. Mamo
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Public Health, Curtin University, Bentley, WA 6102, Australia
| | - Mark J. Hackett
- Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Science, Curtin University, Bentley, WA 6845, Australia
| |
Collapse
|
44
|
Trpm2 Ablation Accelerates Protein Aggregation by Impaired ADPR and Autophagic Clearance in the Brain. Mol Neurobiol 2018; 56:3819-3832. [PMID: 30215158 PMCID: PMC6477016 DOI: 10.1007/s12035-018-1309-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/08/2018] [Indexed: 01/10/2023]
Abstract
TRPM2 a cation channel is also known to work as an enzyme that hydrolyzes highly reactive, neurotoxic ADP-ribose (ADPR). Although ADPR is hydrolyzed by NUT9 pyrophosphatase in major organs, the enzyme is defective in the brain. The present study questions the role of TRPM2 in the catabolism of ADPR in the brain. Genetic ablation of Trpm2 results in the disruption of ADPR catabolism that leads to the accumulation of ADPR and reduction in AMP. Trpm2−/− mice elicit the reduction in autophagosome formation in the hippocampus. Trpm2−/− mice also show aggregations of proteins in the hippocampus, aberrant structural changes and neuronal connections in synapses, and neuronal degeneration. Trpm2−/− mice exhibit learning and memory impairment, enhanced neuronal intrinsic excitability, and imbalanced synaptic transmission. These results respond to long-unanswered questions regarding the potential role of the enzymatic function of TRPM2 in the brain, whose dysfunction evokes protein aggregation. In addition, the present finding answers to the conflicting reports such as neuroprotective or neurodegenerative phenotypes observed in Trpm2−/− mice.
Collapse
|
45
|
Domínguez-González M, Puigpinós M, Jové M, Naudi A, Portero-Otín M, Pamplona R, Ferrer I. Regional vulnerability to lipoxidative damage and inflammation in normal human brain aging. Exp Gerontol 2018; 111:218-228. [PMID: 30077575 DOI: 10.1016/j.exger.2018.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/29/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023]
Abstract
Oxidative damage and inflammation coexist in healthy human brain aging. The present study analyzes levels of protein adduction by lipid peroxidation (LP) end-products neuroketal (NKT) and malondialdehyde (MDA), as markers of protein oxidative damage, cycloxygenase-2 (COX-2) levels, as a marker of inflammation, and cytochrome P450 2J2 (CYP2J2), responsible of generation of neuroprotective products, in twelve brain regions in normal middle-aged individuals (MA) and old-aged (OA) individuals. In addition, levels of these markers were evaluated as a function of age as a continuous variable and correction for multiple comparisons. Selection of regions was based on their different vulnerability to prevalent neurodegenerative diseases in aging. Our findings show region-dependent LP end-products, COX-2 and CYP2J2 changes in the aging human brain. However, no clear relationship can be established between NKT, MDA, COX-2 and CYP2J2 levels, and regional vulnerability to neurodegeneration in old age.
Collapse
Affiliation(s)
- Mayelín Domínguez-González
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain
| | - Meritxell Puigpinós
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain
| | - Mariona Jové
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Alba Naudi
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Manuel Portero-Otín
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida, Biomedical Research Institute of Lleida, UdL-IRBLLeida, 25198 Lleida, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital, Biomedical Research Institute of Bellvitge (IDIBELL), Hospitalet de Llobregat, Spain; Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain; Biomedical Research Network Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain; Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat, Spain.
| |
Collapse
|
46
|
Spagnuolo MS, Bergamo P, Crescenzo R, Iannotta L, Treppiccione L, Iossa S, Cigliano L. Brain Nrf2 pathway, autophagy, and synaptic function proteins are modulated by a short-term fructose feeding in young and adult rats. Nutr Neurosci 2018; 23:309-320. [DOI: 10.1080/1028415x.2018.1501532] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Maria Stefania Spagnuolo
- Department of Bio-Agrofood Science, Institute for the Animal Production System in Mediterranean Environment, National Research Council (CNR-ISPAAM), Naples, Italy
| | - Paolo Bergamo
- Department of Bio-Agrofood Science, Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | | | - Lucia Iannotta
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Lucia Treppiccione
- Department of Bio-Agrofood Science, Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
| | - Susanna Iossa
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Luisa Cigliano
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
47
|
Karri V, Ramos D, Martinez JB, Odena A, Oliveira E, Coort SL, Evelo CT, Mariman ECM, Schuhmacher M, Kumar V. Differential protein expression of hippocampal cells associated with heavy metals (Pb, As, and MeHg) neurotoxicity: Deepening into the molecular mechanism of neurodegenerative diseases. J Proteomics 2018; 187:106-125. [PMID: 30017948 DOI: 10.1016/j.jprot.2018.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 12/29/2022]
Abstract
Chronic exposure to heavy metals such as Pb, As, and MeHg can be associated with an increased risk of developing neurodegenerative diseases. Our in vitro bioassays results showed the potency of heavy metals in the order of Pb < As < MeHg on hippocampal cells. The main objective of this study was combining in vitro label free proteomics and systems biology approach for elucidating patterns of biological response, discovering underlying mechanisms of Pb, As, and MeHg toxicity in hippocampal cells. The omics data was refined by using different filters and normalization and multilevel analysis tools were employed to explore the data visualization. The functional and pathway visualization was performed by using Gene ontology and PathVisio tools. Using these all integrated approaches, we identified significant proteins across treatments within the mitochondrial dysfunction, oxidative stress, ubiquitin proteome dysfunction, and mRNA splicing related to neurodegenerative diseases. The systems biology analysis revealed significant alterations in proteins implicated in Parkinson's disease (PD) and Alzheimer's disease (AD). The current proteomics analysis of three metals support the insight into the proteins involved in neurodegeneration and the altered proteins can be useful for metal-specific biomarkers of exposure and its adverse effects. SIGNIFICANCE The proteomics techniques have been claimed to be more sensitive than the conventional toxicological assays, facilitating the measurement of responses to heavy metals (Pb, As, and MeHg) exposure before obvious harm has occurred demonstrating their predictive value. Also, proteomics allows for the comparison of responses between Pb, As, and MeHg metals, permitting the evaluation of potency differences hippocampal cells of the brain. Hereby, the molecular information provided by pathway and gene functional analysis can be used to develop a more thorough understanding of each metal mechanism at the protein level for different neurological adverse outcomes (e.g. Parkinson's disease, Alzheimer's diseases). Efforts are put into developing proteomics based toxicity testing methods using in vitro models for improving human risk assessment. Some of the key proteins identified can also potentially be used as biomarkers in epidemiologic studies. These heavy metal response patterns shed new light on the mechanisms of mRNA splicing, ubiquitin pathway role in neurodegeneration, and can be useful for the development of molecular biomarkers of heavy metals exposure.
Collapse
Affiliation(s)
- Venkatanaidu Karri
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - David Ramos
- Plataforma de Proteòmica, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Julia Bauzá Martinez
- Plataforma de Proteòmica, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Antonia Odena
- Plataforma de Proteòmica, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Eliandre Oliveira
- Unidad de Toxicologia, Parc Científic de Barcelona, C/Baldiri Reixac, 10-12, 08028 Barcelona, Spain
| | - Susan L Coort
- Department of Bioinformatics, BiGCaT, NUTRIM, Maastricht University, 6229, ER, Maastricht, the Netherlands
| | - Chris T Evelo
- Department of Bioinformatics, BiGCaT, NUTRIM, Maastricht University, 6229, ER, Maastricht, the Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Marta Schuhmacher
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - Vikas Kumar
- Environmental Engineering Laboratory, Departament d'Enginyeria Quimica, Universitat Rovira i Virgili, Av. Països Catalans 26, 43007 Tarragona, Spain; IISPV, Hospital Universitari Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Spain.
| |
Collapse
|
48
|
Moreno-García A, Kun A, Calero O, Medina M, Calero M. An Overview of the Role of Lipofuscin in Age-Related Neurodegeneration. Front Neurosci 2018; 12:464. [PMID: 30026686 PMCID: PMC6041410 DOI: 10.3389/fnins.2018.00464] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/18/2018] [Indexed: 12/21/2022] Open
Abstract
Despite aging being by far the greatest risk factor for highly prevalent neurodegenerative disorders, the molecular underpinnings of age-related brain changes are still not well understood, particularly the transition from normal healthy brain aging to neuropathological aging. Aging is an extremely complex, multifactorial process involving the simultaneous interplay of several processes operating at many levels of the functional organization. The buildup of potentially toxic protein aggregates and their spreading through various brain regions has been identified as a major contributor to these pathologies. One of the most striking morphologic changes in neurons during normal aging is the accumulation of lipofuscin (LF) aggregates, as well as, neuromelanin pigments. LF is an autofluorescent lipopigment formed by lipids, metals and misfolded proteins, which is especially abundant in nerve cells, cardiac muscle cells and skin. Within the Central Nervous System (CNS), LF accumulates as aggregates, delineating a specific senescence pattern in both physiological and pathological states, altering neuronal cytoskeleton and cellular trafficking and metabolism, and being associated with neuronal loss, and glial proliferation and activation. Traditionally, the accumulation of LF in the CNS has been considered a secondary consequence of the aging process, being a mere bystander of the pathological buildup associated with different neurodegenerative disorders. Here, we discuss recent evidence suggesting the possibility that LF aggregates may have an active role in neurodegeneration. We argue that LF is a relevant effector of aging that represents a risk factor or driver for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Alejandra Kun
- Biochemistry Section, Science School, Universidad de la República, Montevideo, Uruguay
- Protein and Nucleic Acids Department, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Olga Calero
- Chronic Disease Programme-CROSADIS, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Miguel Medina
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| | - Miguel Calero
- Chronic Disease Programme-CROSADIS, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
- Alzheimer Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, Madrid, Spain
| |
Collapse
|
49
|
Li P, Hao L, Guo YY, Yang GL, Mei H, Li XH, Zhai QX. Chloroquine inhibits autophagy and deteriorates the mitochondrial dysfunction and apoptosis in hypoxic rat neurons. Life Sci 2018; 202:70-77. [DOI: 10.1016/j.lfs.2018.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 01/28/2023]
|
50
|
Maxwell N, Castro RW, Sutherland NM, Vaughan KL, Szarowicz MD, de Cabo R, Mattison JA, Valdez G. α-Motor neurons are spared from aging while their synaptic inputs degenerate in monkeys and mice. Aging Cell 2018; 17. [PMID: 29397579 PMCID: PMC5847869 DOI: 10.1111/acel.12726] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
Motor function deteriorates with advancing age, increasing the risk of adverse health outcomes. While it is well established that skeletal muscles and neuromuscular junctions (NMJs) degenerate with increasing age, the effect of aging on α‐motor neurons and their innervating synaptic inputs remains largely unknown. In this study, we examined the soma of α‐motor neurons and innervating synaptic inputs in the spinal cord of aged rhesus monkeys and mice, two species with vastly different lifespans. We found that, in both species, α‐motor neurons retain their soma size despite an accumulation of large amounts of cellular waste or lipofuscin. Interestingly, the lipofuscin profile varied considerably, indicating that α‐motor neurons age at different rates. Although the rate of aging varies, α‐motor neurons do not atrophy in old age. In fact, there is no difference in the number of motor axons populating ventral roots in old mice compared to adult mice. Moreover, the transcripts and proteins associated with α‐motor neurons do not decrease in the spinal cord of old mice. However, in aged rhesus monkeys and mice, there were fewer cholinergic and glutamatergic synaptic inputs directly abutting α‐motor neurons, evidence that aging causes α‐motor neurons to shed synaptic inputs. Thus, the loss of synaptic inputs may contribute to age‐related dysfunction of α‐motor neurons. These findings broaden our understanding of the degeneration of the somatic motor system that precipitates motor dysfunction with advancing age.
Collapse
Affiliation(s)
- Nicholas Maxwell
- Virginia Tech Carilion Research Institute; Virginia Tech; Roanoke VA USA
| | - Ryan W. Castro
- Virginia Tech Carilion Research Institute; Virginia Tech; Roanoke VA USA
- Graduate Program in Translational Biology, Medicine, and Health; Virginia Tech; Blacksburg VA USA
| | | | - Kelli L. Vaughan
- Translational Gerontology Branch; National Institute on Aging; NIH; Baltimore MD USA
- SoBran, Inc.; Burtonsville MD USA
| | - Mark D. Szarowicz
- Translational Gerontology Branch; National Institute on Aging; NIH; Baltimore MD USA
- SoBran, Inc.; Burtonsville MD USA
| | - Rafael de Cabo
- Translational Gerontology Branch; National Institute on Aging; NIH; Baltimore MD USA
| | - Julie A. Mattison
- Translational Gerontology Branch; National Institute on Aging; NIH; Baltimore MD USA
| | - Gregorio Valdez
- Virginia Tech Carilion Research Institute; Virginia Tech; Roanoke VA USA
- Department of Biological Sciences; Virginia Tech; Blacksburg VA USA
| |
Collapse
|