1
|
Lucchesi M, Biso L, Bonaso M, Longoni B, Buchignani B, Battini R, Santorelli FM, Doccini S, Scarselli M. Mitochondrial Dysfunction in Genetic and Non-Genetic Parkinson's Disease. Int J Mol Sci 2025; 26:4451. [PMID: 40362688 PMCID: PMC12072996 DOI: 10.3390/ijms26094451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondrial dysfunction is a hallmark of Parkinson's disease (PD) pathogenesis, contributing to increased oxidative stress and impaired endo-lysosomal-proteasome system efficiency underlying neuronal injury. Genetic studies have identified 19 monogenic mutations-accounting for ~10% of PD cases-that affect mitochondrial function and are associated with early- or late-onset PD. Early-onset forms typically involve genes encoding proteins essential for mitochondrial quality control, including mitophagy and structural maintenance, while late-onset mutations impair mitochondrial dynamics, bioenergetics, and trafficking. Atypical juvenile genetic syndromes also exhibit mitochondrial abnormalities. In idiopathic PD, environmental neurotoxins such as pesticides and MPTP act as mitochondrial inhibitors, disrupting complex I activity and increasing reactive oxygen species. These converging pathways underscore mitochondria as a central node in PD pathology. This review explores the overlapping and distinct mitochondrial mechanisms in genetic and non-genetic PD, emphasizing their role in neuronal vulnerability. Targeting mitochondrial dysfunction finally offers a promising therapeutic avenue to slow or modify disease progression by intervening at a key point of neurodegenerative convergence.
Collapse
Affiliation(s)
| | - Letizia Biso
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| | - Marco Bonaso
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| | - Biancamaria Longoni
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| | - Bianca Buchignani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Stefano Doccini
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Marco Scarselli
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| |
Collapse
|
2
|
Ge X, Wekselblatt JB, Elmore S, Wang B, Wang T, Dai R, Zhang T, Dave H, Ghaderi M, Anilkumar AR, Wang B, Sirsi SR, Ahn JM, Shapiro MG, Oka Y, Lois C, Qin Z. In Vivo Cytosolic Delivery of Biomolecules into Neurons for Super-Resolution Imaging and Genome Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501033. [PMID: 40285608 DOI: 10.1002/advs.202501033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Efficient delivery of biomolecules into neurons has significant impacts on therapeutic applications in the central nervous system (CNS) and fundamental neuroscience research. Existing viral and non-viral delivery methods often suffer from inefficient intracellular access due to the endocytic pathway. Here, a neuron-targeting and direct cytosolic delivery platform is discovered by using a 15-amino-acid peptide, termed the N1 peptide, which enables neuron-specific targeting and cytosolic delivery of functional biomolecules. The N1 peptide initially binds hyaluronan in the extracellular matrix and subsequently passes the membrane of neurons without being trapped into endosome. This mechanism facilitates the efficient delivery of cell-impermeable and photo-stable fluorescent dye for super-resolution imaging of dendritic spines, and functional proteins, such as Cre recombinase, for site-specific genome modification. Importantly, the N1 peptide exhibits robust neuronal specificity across diverse species, including mice, rats, tree shrews, and zebra finches. Its targeting capability is further demonstrated through various administration routes, including intraparenchymal, intrathecal, and intravenous (i.v.) injections after blood-brain barrier (BBB) opening with focused ultrasound (FUS). These findings establish the N1 peptide as a versatile and functional platform with significant potential for bioimaging and advanced therapeutic applications.
Collapse
Affiliation(s)
- Xiaoqian Ge
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Joseph B Wekselblatt
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Department of Opthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Scott Elmore
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Bo Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tongtong Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Renjinming Dai
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Tingting Zhang
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Harsh Dave
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Mohammadaref Ghaderi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Athul Raj Anilkumar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Bill Wang
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Shashank R Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Jung-Mo Ahn
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Howard Hughes Medical Institute, Pasadena, CA, 91125, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Zhenpeng Qin
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080-3021, USA
| |
Collapse
|
3
|
Nibrad D, Shiwal A, Tadas M, Katariya R, Kale M, Kotagale N, Umekar M, Taksande B. Therapeutic modulation of mitochondrial dynamics by agmatine in neurodegenerative disorders. Neuroscience 2025; 569:43-57. [PMID: 39890051 DOI: 10.1016/j.neuroscience.2025.01.061] [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: 12/10/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Mitochondrial dysfunction is a pivotal factor in the pathogenesis of neurodegenerative disorders, driving neuronal degeneration through mechanisms involving oxidative stress, impaired energy production, and dysregulated calcium homeostasis. Agmatine, an endogenous polyamine derived from arginine, has garnered attention for its neuroprotective properties, including anti-inflammatory, anti-oxidative, and antiapoptotic effects. Recent studies have highlighted the potential of agmatine in preserving mitochondrial function and mitigating neurodegeneration, making it a promising candidate for therapeutic intervention. One of the key mechanisms by which agmatine exerts its neuroprotective effects is through the maintenance of mitochondrial homeostasis. Agmatine has been shown to modulate mitochondrial dynamics, promoting mitochondrial fusion and fission balance essential for cellular energy metabolism and signaling. Moreover, agmatine acts as a regulator of mitochondrial permeability transition pore (mPTP) opening, preventing excessive calcium influx and subsequent mitochondrial dysfunction. Despite promising findings, challenges such as optimizing agmatine's pharmacokinetics, determining optimal dosing regimens, and elucidating its precise molecular targets within mitochondria remain to be addressed. Future research directions should focus on developing targeted delivery systems for agmatine, investigating its interactions with mitochondrial proteins, and conducting well-designed clinical trials to evaluate its therapeutic efficacy and safety profile in neurodegenerative disorders. Overall, agmatine emerges as a novel therapeutic agent with the potential to modulate mitochondrial homeostasis and alleviate neurodegenerative pathology, offering new avenues for treating these debilitating conditions.
Collapse
Affiliation(s)
- Dhanshree Nibrad
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Amit Shiwal
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Manasi Tadas
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Raj Katariya
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Mayur Kale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, (M.S.) 444604, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, (M.S.) 441 002, India.
| |
Collapse
|
4
|
Tournezy J, Léger C, Klonjkowski B, Gonzalez-Dunia D, Szelechowski M, Garenne A, Mathis S, Chevallier S, Le Masson G. The Neuroprotective Effect of the X Protein of Orthobornavirus Bornaense Type 1 in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2024; 25:12789. [PMID: 39684507 DOI: 10.3390/ijms252312789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
In amyotrophic lateral sclerosis (ALS), early mitochondrial dysfunction may contribute to progressive motor neuron loss. Remarkably, the ectopic expression of the Orthobornavirus bornaense type 1 (BoDV-1) X protein in mitochondria blocks apoptosis and protects neurons from degeneration. Therefore, this study examines the neuroprotective effects of X protein in an ALS mouse model. We first tested in vitro the effect of the X-derived peptide (PX3) on motoneurons primary cultures of SOD1G93A mice. The total intracellular adenosine triphosphate (ATP) content was measured after incubation of the peptide. We next tested in vivo the intramuscular injection of X protein using a canine viral vector (CAV2-X) and PX3 intranasal administrations in SOD1G93A mice. Disease onset and progression were assessed through rotarod performance, functional motor unit analysis via electrophysiology, and motor neuron survival by immunohistochemistry. The results showed that in vitro PX3 restored the ATP level in SOD1G93A motor neurons. In vivo, treated mice demonstrated better motor performance, preserved motor units, and higher motor neuron survival. Although life expectancy was not extended in this severe mouse model of motor neuron degeneration, the present findings clearly demonstrate the neuroprotective potential of X protein in a model of ALS. We are convinced that further studies may improve the therapeutic impact of X protein with optimized administration methods.
Collapse
Affiliation(s)
- Jeflie Tournezy
- Neurocentre Magendie INSERM U1215, Université de Bordeaux, 33000 Bordeaux, France
| | - Claire Léger
- Neurocentre Magendie INSERM U1215, Université de Bordeaux, 33000 Bordeaux, France
| | - Bernard Klonjkowski
- UMR 1161 Virologie, INRA, ANSES, Ecole Nationale Vétérinaire d'Alfort, 94700 Maisons-Alfort, France
| | - Daniel Gonzalez-Dunia
- Infinity (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM, CNRS, Université de Toulouse, UPS, 31024 Toulouse, France
| | - Marion Szelechowski
- Infinity (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM, CNRS, Université de Toulouse, UPS, 31024 Toulouse, France
| | - André Garenne
- IMS Laboratory, UMR5218, CNRS, Bordeaux University, 33400 Talence, France
| | - Stéphane Mathis
- Nerve-Muscle Unit, ALS Center, Department of Neurology, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), 33000 Bordeaux, France
| | - Stéphanie Chevallier
- Neurocentre Magendie INSERM U1215, Université de Bordeaux, 33000 Bordeaux, France
| | - Gwendal Le Masson
- Neurocentre Magendie INSERM U1215, Université de Bordeaux, 33000 Bordeaux, France
- Nerve-Muscle Unit, ALS Center, Department of Neurology, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), 33000 Bordeaux, France
| |
Collapse
|
5
|
Osei D, Baumgart-Vogt E, Ahlemeyer B, Herden C. Tumor Necrosis Factor-α Receptor 1 Mediates Borna Disease Virus 1-Induced Changes in Peroxisomal and Mitochondrial Dynamics in Neurons. Int J Mol Sci 2024; 25:1849. [PMID: 38339126 PMCID: PMC10855776 DOI: 10.3390/ijms25031849] [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: 12/23/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Borna disease virus 1 (BoDV1) causes a persistent infection in the mammalian brain. Peroxisomes and mitochondria play essential roles in the cellular antiviral immune response, but the effect of BoDV1 infection on peroxisomal and mitochondrial dynamics and their respective antioxidant capacities is still not clear. Using different mouse lines-i.e., tumor necrosis factor-α transgenic (TNFTg; to pro-inflammatory status), TNF receptor-1 knockout (TNFR1ko), and TNFR2ko mice in comparison to wild-type (Wt) mice-we analyzed the abundances of both organelles and their main antioxidant enzymes, catalase and superoxide dismutase 2 (SOD2), in neurons of the hippocampal, cerebral, and cerebellar cortices. In TNFTg mice, a strong increase in mitochondrial (6.9-fold) and SOD2 (12.1-fold) abundances was detected; meanwhile, peroxisomal abundance increased slightly (1.5-fold), but that of catalase decreased (2.9-fold). After BoDV1 infection, a strong decrease in mitochondrial (2.1-6.5-fold), SOD2 (2.7-9.1-fold), and catalase (2.7-10.3-fold) abundances, but a slight increase in peroxisomes (1.3-1.6-fold), were detected in Wt and TNFR2ko mice, whereas no changes occurred in TNFR1ko mice. Our data suggest that the TNF system plays a crucial role in the biogenesis of both subcellular organelles. Moreover, TNFR1 signaling mediated the changes in peroxisomal and mitochondrial dynamics after BoDV1 infection, highlighting new mechanisms by which BoDV1 may achieve immune evasion and viral persistence.
Collapse
Affiliation(s)
- Dominic Osei
- Institute for Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; (D.O.); (E.B.-V.)
- Institute of Veterinary Pathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; (D.O.); (E.B.-V.)
| | - Barbara Ahlemeyer
- Institute for Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; (D.O.); (E.B.-V.)
| | - Christiane Herden
- Institute of Veterinary Pathology, Justus Liebig University Giessen, 35392 Giessen, Germany
- Center for Mind, Brain and Behavior, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
6
|
Theme 07 - Pre-Clinical Therapeutic Strategies. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:173-191. [PMID: 37966323 DOI: 10.1080/21678421.2023.2260198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
7
|
Abrishamdar M, Jalali MS, Farbood Y. Targeting Mitochondria as a Therapeutic Approach for Parkinson's Disease. Cell Mol Neurobiol 2023; 43:1499-1518. [PMID: 35951210 PMCID: PMC11412433 DOI: 10.1007/s10571-022-01265-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
Neurodegeneration is among the most critical challenges that involve modern societies and annually influences millions of patients worldwide. While the pathophysiology of Parkinson's disease (PD) is complicated, the role of mitochondrial is demonstrated. The in vitro and in vivo models and genome-wide association studies in human cases proved that specific genes, including PINK1, Parkin, DJ-1, SNCA, and LRRK2, linked mitochondrial dysfunction with PD. Also, mitochondrial DNA (mtDNA) plays an essential role in the pathophysiology of PD. Targeting mitochondria as a therapeutic approach to inhibit or slow down PD formation and progression seems to be an exciting issue. The current review summarized known mutations associated with both mitochondrial dysfunction and PD. The significance of mtDNA in Parkinson's disease pathogenesis and potential PD therapeutic approaches targeting mitochondrial dysfunction was then discussed.
Collapse
Affiliation(s)
- Maryam Abrishamdar
- Department of Physiology, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Sadat Jalali
- Department of Physiology, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Yaghoob Farbood
- Department of Physiology, Medicine Faculty, Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Modulation of mitochondria by viral proteins. Life Sci 2023; 313:121271. [PMID: 36526048 DOI: 10.1016/j.lfs.2022.121271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/24/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022]
Abstract
Mitochondria are dynamic cellular organelles with diverse functions including energy production, calcium homeostasis, apoptosis, host innate immune signaling, and disease progression. Several viral proteins specifically target mitochondria to subvert host defense as mitochondria stand out as the most suitable target for the invading viruses. They have acquired the capability to control apoptosis, metabolic state, and evade immune responses in host cells, by targeting mitochondria. In this way, the viruses successfully allow the spread of viral progeny and thus the infection. Viruses employ their proteins to alter mitochondrial dynamics and their specific functions by a modulation of membrane potential, reactive oxygen species, calcium homeostasis, and mitochondrial bioenergetics to help them achieve a state of persistent infection. A better understanding of such viral proteins and their impact on mitochondrial forms and functions is the main focus of this review. We also attempt to emphasize the importance of exploring the role of mitochondria in the context of SARS-CoV2 pathogenesis and identify host-virus protein interactions.
Collapse
|
9
|
Kanda T, Tomonaga K. Reverse Genetics and Artificial Replication Systems of Borna Disease Virus 1. Viruses 2022; 14:v14102236. [PMID: 36298790 PMCID: PMC9612284 DOI: 10.3390/v14102236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
Borna disease virus 1 (BoDV-1) is a neurotropic RNA virus belonging to the family Bornaviridae within the order Mononegavirales. Whereas BoDV-1 causes neurological and behavioral disorders, called Borna disease (BD), in a wide range of mammals, its virulence in humans has been debated for several decades. However, a series of case reports in recent years have established the nature of BoDV-1 as a zoonotic pathogen that causes fatal encephalitis in humans. Although many virological properties of BoDV-1 have been revealed to date, the mechanism by which it causes fatal encephalitis in humans remains unclear. In addition, there are no effective vaccines or antiviral drugs that can be used in clinical practice. A reverse genetics approach to generating replication-competent recombinant viruses from full-length cDNA clones is a powerful tool that can be used to not only understand viral properties but also to develop vaccines and antiviral drugs. The rescue of recombinant BoDV-1 (rBoDV-1) was first reported in 2005. However, due to the slow nature of the replication of this virus, the rescue of high-titer rBoDV-1 required several months, limiting the use of this system. This review summarizes the history of the reverse genetics and artificial replication systems for orthobornaviruses and explores the recent progress in efforts to rescue rBoDV-1.
Collapse
Affiliation(s)
- Takehiro Kanda
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Molecular Virology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Keizo Tomonaga
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Molecular Virology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto 606-8507, Japan
- Correspondence:
| |
Collapse
|
10
|
Leggio L, L'Episcopo F, Magrì A, Ulloa‐Navas MJ, Paternò G, Vivarelli S, Bastos CAP, Tirolo C, Testa N, Caniglia S, Risiglione P, Pappalardo F, Serra A, García‐Tárraga P, Faria N, Powell JJ, Peruzzotti‐Jametti L, Pluchino S, García‐Verdugo JM, Messina A, Marchetti B, Iraci N. Small Extracellular Vesicles Secreted by Nigrostriatal Astrocytes Rescue Cell Death and Preserve Mitochondrial Function in Parkinson's Disease. Adv Healthc Mater 2022; 11:e2201203. [PMID: 35856921 PMCID: PMC11468249 DOI: 10.1002/adhm.202201203] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/05/2022] [Indexed: 01/28/2023]
Abstract
Extracellular vesicles (EVs) are emerging as powerful players in cell-to-cell communication both in healthy and diseased brain. In Parkinson's disease (PD)-characterized by selective dopaminergic neuron death in ventral midbrain (VMB) and degeneration of their terminals in striatum (STR)-astrocytes exert dual harmful/protective functions, with mechanisms not fully elucidated. Here, this study shows that astrocytes from the VMB-, STR-, and VMB/STR-depleted brains release a population of small EVs in a region-specific manner. Interestingly, VMB-astrocytes secreted the highest rate of EVs, which is further exclusively increased in response to CCL3, a chemokine that promotes robust dopaminergic neuroprotection in different PD models. The neuroprotective potential of nigrostriatal astrocyte-EVs is investigated in differentiated versus undifferentiated SH-SY5Y cells exposed to oxidative stress and mitochondrial toxicity. EVs from both VMB- and STR-astrocytes counteract H2 O2 -induced caspase-3 activation specifically in differentiated cells, with EVs from CCL3-treated astrocytes showing a higher protective effect. High resolution respirometry further reveals that nigrostriatal astrocyte-EVs rescue neuronal mitochondrial complex I function impaired by the neurotoxin MPP+ . Notably, only EVs from VMB-astrocyte fully restore ATP production, again specifically in differentiated SH-SY5Y. These results highlight a regional diversity in the nigrostriatal system for the secretion and activities of astrocyte-EVs, with neuroprotective implications for PD.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | - Andrea Magrì
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - María José Ulloa‐Navas
- Laboratory of Compared NeurobiologyUniversity of Valencia‐CIBERNEDPaterna46980Spain
- Department of NeuroscienceMayo ClinicJacksonvilleFL32257USA
| | - Greta Paternò
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | | | | | | | - Pierpaolo Risiglione
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - Fabrizio Pappalardo
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| | | | | | - Nuno Faria
- Department of Veterinary MedicineUniversity of CambridgeCambridgeCB3 0ESUK
| | - Jonathan J. Powell
- Department of Veterinary MedicineUniversity of CambridgeCambridgeCB3 0ESUK
| | | | - Stefano Pluchino
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | | | - Angela Messina
- Department of Biological, Geological and Environmental SciencesUniversity of CataniaCatania95125Italy
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
- Oasi Research Institute‐IRCCSTroina94018Italy
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCatania95123Italy
| |
Collapse
|
11
|
Wang Y, Qian S, Zhao F, Wang Y, Li J. Terazosin Analogs Targeting Pgk1 as Neuroprotective Agents: Design, Synthesis, and Evaluation. Front Chem 2022; 10:906974. [PMID: 35958233 PMCID: PMC9360532 DOI: 10.3389/fchem.2022.906974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Nitrogen-containing heterocyclic compounds have shown promising therapeutic effects in a variety of inflammatory and neurodegenerative diseases. Recently, terazosin (TZ), a heterocyclic compound with a quinazoline core, was found to combine with phosphoglycerol kinase 1 (Pgk1) and protect neurons by enhancing Pgk1 activity and promoting glycolysis, thereby slowing, or preventing the neurodegeneration of PD. These findings indicated that terazosin analogs have bright prospects for the development of PD therapeutics. In this study, a series of terazosin analogs were designed and synthesized for neuroprotective effects by targeting Pgk1. Among them, compound 12b was obtained with the best Pgk1 agonistic activity and neuroprotective activity. Further study indicates that it can increase intracellular ATP content and reduce ROS levels by stimulating the activity of Pgk1, thereby playing a role in protecting nerve cells. In conclusion, this study provides a new strategy and reference for the development of neuroprotective drugs.
Collapse
Affiliation(s)
- Yang Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, China
| | - Shihu Qian
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, China
| | - Fang Zhao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, China
| | - Yujie Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, China
| | - Jiaming Li
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, China
- *Correspondence: Jiaming Li,
| |
Collapse
|
12
|
Rubbenstroth D. Avian Bornavirus Research—A Comprehensive Review. Viruses 2022; 14:v14071513. [PMID: 35891493 PMCID: PMC9321243 DOI: 10.3390/v14071513] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
Avian bornaviruses constitute a genetically diverse group of at least 15 viruses belonging to the genus Orthobornavirus within the family Bornaviridae. After the discovery of the first avian bornaviruses in diseased psittacines in 2008, further viruses have been detected in passerines and aquatic birds. Parrot bornaviruses (PaBVs) possess the highest veterinary relevance amongst the avian bornaviruses as the causative agents of proventricular dilatation disease (PDD). PDD is a chronic and often fatal disease that may engulf a broad range of clinical presentations, typically including neurologic signs as well as impaired gastrointestinal motility, leading to proventricular dilatation. It occurs worldwide in captive psittacine populations and threatens private bird collections, zoological gardens and rehabilitation projects of endangered species. In contrast, only little is known about the pathogenic roles of passerine and waterbird bornaviruses. This comprehensive review summarizes the current knowledge on avian bornavirus infections, including their taxonomy, pathogenesis of associated diseases, epidemiology, diagnostic strategies and recent developments on prophylactic and therapeutic countermeasures.
Collapse
Affiliation(s)
- Dennis Rubbenstroth
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald, Insel Riems, Germany
| |
Collapse
|
13
|
HSPA9/Mortalin mediates axo-protection and modulates mitochondrial dynamics in neurons. Sci Rep 2021; 11:17705. [PMID: 34489498 PMCID: PMC8421332 DOI: 10.1038/s41598-021-97162-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 08/23/2021] [Indexed: 12/21/2022] Open
Abstract
Mortalin is a mitochondrial chaperone protein involved in quality control of proteins imported into the mitochondrial matrix, which was recently described as a sensor of neuronal stress. Mortalin is down-regulated in neurons of patients with neurodegenerative diseases and levels of Mortalin expression are correlated with neuronal fate in animal models of Alzheimer's disease or cerebral ischemia. To date, however, the links between Mortalin levels, its impact on mitochondrial function and morphology and, ultimately, the initiation of neurodegeneration, are still unclear. In the present study, we used lentiviral vectors to over- or under-express Mortalin in primary neuronal cultures. We first analyzed the early events of neurodegeneration in the axonal compartment, using oriented neuronal cultures grown in microfluidic-based devices. We observed that Mortalin down-regulation induced mitochondrial fragmentation and axonal damage, whereas its over-expression conferred protection against axonal degeneration mediated by rotenone exposure. We next demonstrated that Mortalin levels modulated mitochondrial morphology by acting on DRP1 phosphorylation, thereby further illustrating the crucial implication of mitochondrial dynamics on neuronal fate in degenerative diseases.
Collapse
|
14
|
Pavesi A. Origin, Evolution and Stability of Overlapping Genes in Viruses: A Systematic Review. Genes (Basel) 2021; 12:genes12060809. [PMID: 34073395 PMCID: PMC8227390 DOI: 10.3390/genes12060809] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022] Open
Abstract
During their long evolutionary history viruses generated many proteins de novo by a mechanism called “overprinting”. Overprinting is a process in which critical nucleotide substitutions in a pre-existing gene can induce the expression of a novel protein by translation of an alternative open reading frame (ORF). Overlapping genes represent an intriguing example of adaptive conflict, because they simultaneously encode two proteins whose freedom to change is constrained by each other. However, overlapping genes are also a source of genetic novelties, as the constraints under which alternative ORFs evolve can give rise to proteins with unusual sequence properties, most importantly the potential for novel functions. Starting with the discovery of overlapping genes in phages infecting Escherichia coli, this review covers a range of studies dealing with detection of overlapping genes in small eukaryotic viruses (genomic length below 30 kb) and recognition of their critical role in the evolution of pathogenicity. Origin of overlapping genes, what factors favor their birth and retention, and how they manage their inherent adaptive conflict are extensively reviewed. Special attention is paid to the assembly of overlapping genes into ad hoc databases, suitable for future studies, and to the development of statistical methods for exploring viral genome sequences in search of undiscovered overlaps.
Collapse
Affiliation(s)
- Angelo Pavesi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 23/A, I-43124 Parma, Italy
| |
Collapse
|
15
|
Zhang W, Chen H, Ding L, Gong J, Zhang M, Guo W, Xu P, Li S, Zhang Y. Trojan Horse Delivery of 4,4'-Dimethoxychalcone for Parkinsonian Neuroprotection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004555. [PMID: 33977069 PMCID: PMC8097374 DOI: 10.1002/advs.202004555] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/05/2021] [Indexed: 05/04/2023]
Abstract
Parkinson's disease (PD) is characterized by the progressive deterioration of dopamine (DA) neurons, and therapeutic endeavors are aimed at preventing DA loss. However, lack of effective brain delivery approaches limits this strategy. In this study, a "Trojan horse" system is used for substantia nigra-targeted delivery of a blood brain barrier-penetrating peptide (RVG29) conjugated to the surface of nanoparticles loaded with the natural autophagy inducer 4,4'-dimethoxychalcone (DMC) (designated as RVG-nDMC). Here, the neuroprotective effects of DMC are demonstrated in PD. Specifically, RVG-nDMC penetrates the blood brain barrier with enhanced brain-targeted delivery efficiency and is internalized by DA neurons and microglia. In vivo studies demonstrate that RVG-nDMC ameliorates motor deficits and nigral DA neuron death in PD mice without causing overt adverse effects in the brain or other major organs. Moreover, RVG-nDMC reverses tyrosine hydroxylase ubiquitination and degradation, alleviates oxidative stress in DA neurons, and exerts antiinflammatory effects in microglia. The "Trojan horse" strategy for targeted delivery of DMC thus provides a potentially powerful and clinically feasible approach for PD intervention.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of NeurologyThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510120China
| | - Huaqing Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhou511436China
| | - Liuyan Ding
- Department of NeurologyThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510120China
| | - Junwei Gong
- Key Laboratory of Neurological Function and HealthSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Mengran Zhang
- Key Laboratory of Neurological Function and HealthSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Wenyuan Guo
- Department of NeurologyThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510120China
| | - Pingyi Xu
- Department of NeurologyThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510120China
| | - Shiying Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences & The Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhou511436China
| | - Yunlong Zhang
- Key Laboratory of Neurological Function and HealthSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| |
Collapse
|
16
|
Alsharif I, Boukhzar L, Lefranc B, Godefroy D, Aury-Landas J, Rego JLD, Rego JCD, Naudet F, Arabo A, Chagraoui A, Maltête D, Benazzouz A, Baugé C, Leprince J, Elkahloun AG, Eiden LE, Anouar Y. Cell-penetrating, antioxidant SELENOT mimetic protects dopaminergic neurons and ameliorates motor dysfunction in Parkinson's disease animal models. Redox Biol 2021; 40:101839. [PMID: 33486153 PMCID: PMC7823055 DOI: 10.1016/j.redox.2020.101839] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor dysfunction for which there is an unmet need for better treatment options. Although oxidative stress is a common feature of neurodegenerative diseases, notably PD, there is currently no efficient therapeutic strategy able to tackle this multi-target pathophysiological process. Based on our previous observations of the potent antioxidant and neuroprotective activity of SELENOT, a vital thioredoxin-like selenoprotein, we designed the small peptide PSELT from its redox active site to evaluate its antioxidant properties in vivo, and its potential polyfunctional activity in PD models. PSELT protects neurotoxin-treated dopaminergic neurons against oxidative stress and cell death, and their fibers against neurotoxic degeneration. PSELT is cell-permeable and acts in multiple subcellular compartments of dopaminergic neurons that are vulnerable to oxidative stress. In rodent models of PD, this protective activity prevented neurodegeneration, restored phosphorylated tyrosine hydroxylase levels, and led to improved motor skills. Transcriptomic analysis revealed that gene regulation by PSELT after MPP+ treatment negatively correlates with that occurring in PD, and positively correlates with that occurring after resveratrol treatment. Mechanistically, a major impact of PSELT is via nuclear stimulation of the transcription factor EZH2, leading to neuroprotection. Overall, these findings demonstrate the potential of PSELT as a therapeutic candidate for treatment of PD, targeting oxidative stress at multiple intracellular levels.
Collapse
Affiliation(s)
- Ifat Alsharif
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France; Biology department, Jamoum University College, Umm Alqura University, Saudi Arabia
| | - Loubna Boukhzar
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France
| | - Benjamin Lefranc
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France; PRIMACEN, Cell Imaging Platform of Normandie, UNIROUEN, 76000, Rouen, France
| | - David Godefroy
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France
| | | | - Jean-Luc do Rego
- Institute for Research and Innovation in Biomedicine, 76000, Rouen, France; Behavioral Analysis Platform SCAC, Rouen Medical School, Rouen Normandie University, 76183, Rouen, France
| | - Jean-Claude do Rego
- Institute for Research and Innovation in Biomedicine, 76000, Rouen, France; Behavioral Analysis Platform SCAC, Rouen Medical School, Rouen Normandie University, 76183, Rouen, France
| | - Frédéric Naudet
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, Bordeaux University, F-33000, Bordeaux, France
| | - Arnaud Arabo
- Biological Resource Service (SRB), Faculty of Sciences and Techniques, Rouen Normandie University, 76821, Mont-Saint-Aignan, France
| | - Abdeslam Chagraoui
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France
| | - David Maltête
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France
| | - Abdelhamid Benazzouz
- Institut des Maladies Neurodégénératives, CNRS, UMR 5293, Bordeaux University, F-33000, Bordeaux, France
| | | | - Jérôme Leprince
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France; PRIMACEN, Cell Imaging Platform of Normandie, UNIROUEN, 76000, Rouen, France
| | - Abdel G Elkahloun
- Comparative Genomics and Cancer, Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Youssef Anouar
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen Normandie University, 76821, Mont-Saint-Aignan, France; Institute for Research and Innovation in Biomedicine, 76000, Rouen, France.
| |
Collapse
|
17
|
Nobach D, Müller J, Tappe D, Herden C. Update on immunopathology of bornavirus infections in humans and animals. Adv Virus Res 2020; 107:159-222. [PMID: 32711729 DOI: 10.1016/bs.aivir.2020.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Knowledge on bornaviruses has expanded tremendously during the last decade through detection of novel bornaviruses and endogenous bornavirus-like elements in many eukaryote genomes, as well as by confirmation of insectivores as reservoir species for classical Borna disease virus 1 (BoDV-1). The most intriguing finding was the demonstration of the zoonotic potential of lethal human bornavirus infections caused by a novel bornavirus of different squirrel species (variegated squirrel 1 bornavirus, VSBV-1) and by BoDV-1 known as the causative agent for the classical Borna disease in horses and sheep. Whereas a T cell-mediated immunopathology has already been confirmed as key disease mechanism for infection with BoDV-1 by experimental studies in rodents, the underlying pathomechanisms remain less clear for human bornavirus infections, infection with other bornaviruses or infection of reservoir species. Thus, an overview of current knowledge on the pathogenesis of bornavirus infections focusing on immunopathology is given.
Collapse
Affiliation(s)
- Daniel Nobach
- Institute of Veterinary Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jana Müller
- Institute of Veterinary Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Christiane Herden
- Institute of Veterinary Pathology, Justus-Liebig-University Giessen, Giessen, Germany; Center for Brain, Mind and Behavior, Justus-Liebig-University Giessen, Giessen, Germany.
| |
Collapse
|
18
|
Scorziello A, Borzacchiello D, Sisalli MJ, Di Martino R, Morelli M, Feliciello A. Mitochondrial Homeostasis and Signaling in Parkinson's Disease. Front Aging Neurosci 2020; 12:100. [PMID: 32372945 PMCID: PMC7186467 DOI: 10.3389/fnagi.2020.00100] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
The loss of dopaminergic (DA) neurons in the substantia nigra leads to a progressive, long-term decline of movement and other non-motor deficits. The symptoms of Parkinson's disease (PD) often appear later in the course of the disease, when most of the functional dopaminergic neurons have been lost. The late onset of the disease, the severity of the illness, and its impact on the global health system demand earlier diagnosis and better targeted therapy. PD etiology and pathogenesis are largely unknown. There are mutations in genes that have been linked to PD and, from these complex phenotypes, mitochondrial dysfunction emerged as central in the pathogenesis and evolution of PD. In fact, several PD-associated genes negatively impact on mitochondria physiology, supporting the notion that dysregulation of mitochondrial signaling and homeostasis is pathogenically relevant. Derangement of mitochondrial homeostatic controls can lead to oxidative stress and neuronal cell death. Restoring deranged signaling cascades to and from mitochondria in PD neurons may then represent a viable opportunity to reset energy metabolism and delay the death of dopaminergic neurons. Here, we will highlight the relevance of dysfunctional mitochondrial homeostasis and signaling in PD, the molecular mechanisms involved, and potential therapeutic approaches to restore mitochondrial activities in damaged neurons.
Collapse
Affiliation(s)
- Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maria Jose Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Rossana Di Martino
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
19
|
Pavesi A. New insights into the evolutionary features of viral overlapping genes by discriminant analysis. Virology 2020; 546:51-66. [PMID: 32452417 PMCID: PMC7157939 DOI: 10.1016/j.virol.2020.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
Abstract
Overlapping genes originate by a mechanism of overprinting, in which nucleotide substitutions in a pre-existing frame induce the expression of a de novo protein from an alternative frame. In this study, I assembled a dataset of 319 viral overlapping genes, which included 82 overlaps whose expression is experimentally known and the respective 237 homologs. Principal component analysis revealed that overlapping genes have a common pattern of nucleotide and amino acid composition. Discriminant analysis separated overlapping from non-overlapping genes with an accuracy of 97%. When applied to overlapping genes with known genealogy, it separated ancestral from de novo frames with an accuracy close to 100%. This high discriminant power was crucial to computationally design variants of de novo viral proteins known to possess selective anticancer toxicity (apoptin) or protection against neurodegeneration (X protein), as well as to detect two new potential overlapping genes in the genome of the new coronavirus SARS-CoV-2.
Collapse
Affiliation(s)
- Angelo Pavesi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area Delle Scienze 23/A, I-43124, Parma, Italy.
| |
Collapse
|
20
|
Liu J, Liu C, Zhang J, Zhang Y, Liu K, Song JX, Sreenivasmurthy SG, Wang Z, Shi Y, Chu C, Zhang Y, Wu C, Deng X, Liu X, Song J, Zhuang R, Huang S, Zhang P, Li M, Wen L, Zhang YW, Liu G. A Self-Assembled α-Synuclein Nanoscavenger for Parkinson's Disease. ACS NANO 2020; 14:1533-1549. [PMID: 32027482 DOI: 10.1021/acsnano.9b06453] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Although emerging evidence suggests that the pathogenesis of Parkinson's disease (PD) is closely related to the aggregation of alpha-synuclein (α-syn) in the midbrain, the clearance of α-syn remains an unmet clinical need. Here, we develop a simple and efficient strategy for fabricating the α-syn nanoscavenger for PD via a reprecipitation self-assembly procedure. The curcumin analogue-based nanoscavenger (NanoCA) is engineered to be capable of a controlled-release property to stimulate nuclear translocation of the major autophagy regulator, transcription factor EB (TFEB), triggering both autophagy and calcium-dependent exosome secretion for the clearance of α-syn. Pretreatment of NanoCA protects cell lines and primary neurons from MPP+-induced neurotoxicity. More importantly, a rapid arousal intranasal delivery system (RA-IDDS) was designed and applied for the brain-targeted delivery of NanoCA, which affords robust neuroprotection against behavioral deficits and promotes clearance of monomer, oligomer, and aggregates of α-syn in the midbrain of an MPTP mouse model of PD. Our findings provide a clinically translatable therapeutic strategy aimed at neuroprotection and disease modification in PD.
Collapse
Affiliation(s)
- Jingyi Liu
- School of Medicine , Xiamen University , Xiamen 361102 , China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine , Hong Kong Baptist University , Hong Kong SAR , China
| | - Chao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences , Beijing Institute of Technology , Beijing 100081 , China
| | - Yunming Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Keyin Liu
- School of Medicine , Xiamen University , Xiamen 361102 , China
| | - Ju-Xian Song
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine , Hong Kong Baptist University , Hong Kong SAR , China
| | | | - Ziying Wang
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine , Hong Kong Baptist University , Hong Kong SAR , China
| | - Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Chengchao Chu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Caisheng Wu
- Laboratory Animal Center , Xiamen University , Xiamen 361102 , China
- School of Pharmaceutical Sciences , Xiamen University , Xiamen 361102 , China
| | - Xianhua Deng
- School of Medicine , Xiamen University , Xiamen 361102 , China
| | - Xingyang Liu
- School of Medicine , Xiamen University , Xiamen 361102 , China
| | - Jing Song
- Laboratory Animal Center , Xiamen University , Xiamen 361102 , China
- School of Pharmaceutical Sciences , Xiamen University , Xiamen 361102 , China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Shuqiong Huang
- School of Medicine , Xiamen University , Xiamen 361102 , China
| | - Pengfei Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine , Hong Kong Baptist University , Hong Kong SAR , China
| | - Lei Wen
- School of Medicine , Xiamen University , Xiamen 361102 , China
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience , Xiamen University , Xiamen 361102 , China
| | - Yun Wu Zhang
- School of Medicine , Xiamen University , Xiamen 361102 , China
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience , Xiamen University , Xiamen 361102 , China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , China
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience , Xiamen University , Xiamen 361102 , China
| |
Collapse
|
21
|
Geng J, Liu W, Gao J, Jiang C, Fan T, Sun Y, Qin Z, Xu Q, Guo W, Gao J. Andrographolide alleviates Parkinsonism in MPTP-PD mice via targeting mitochondrial fission mediated by dynamin-related protein 1. Br J Pharmacol 2019; 176:4574-4591. [PMID: 31389613 PMCID: PMC6932945 DOI: 10.1111/bph.14823] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 07/17/2019] [Accepted: 07/20/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Accumulating evidence indicates that mitochondrial dynamics play an important role in the progressive deterioration of dopaminergic neurons. Andrographolide has been found to exert neuroprotective effects in several models of neurological diseases. However, the mechanism of how andrographolide protects neurons in Parkinson's disease (PD) remains not fully understood. EXPERIMENTAL APPROACH Behavioural experiments were performed to examine the effect of andrographolide in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-PD mice. Mitochondrial mass and morphology were visualized using transmission electron microscopy (TEM). SH-SY5Y cells and primary mouse neurons were exposed to rotenone to mimic PD in vitro. Western blotting, co-immunoprecipitation and immunofluorescence were performed. The target protein of andrographolide was identified by biotin-andrographolide pulldown assay as well as drug affinity responsive target stability (DARTS), cellular thermal shift (CETSA), and surface plasmon resonance (SPR) assays. KEY RESULTS Andrographolide administration improved behavioural deficits and attenuated loss of dopaminergic neurons in MPTP-exposed mice and reduced cell death induced by rotenone in vitro. An increased mitochondrial mass, and decreased surface area were found in the striatum from MPTP-PD mice, as well as in rotenone-treated primary neurons and SH-SY5Y cells, while andrographolide treatment preserved mitochondrial mass and morphology. Dynamin-related protein 1 (DRP1) was identified as a target protein of andrographolide. Andrographolide bound to DRP1 and inhibited its GTPase activity, thereby preventing excessive mitochondria fission and neuronal damage in PD. CONCLUSIONS AND IMPLICATIONS Our findings suggest that andrographolide may protect neurons against rotenone- or MPTP-induced damage in vitro and in vivo through inhibiting mitochondrial fission.
Collapse
Affiliation(s)
- Ji Geng
- School of PharmacyJiangsu UniversityZhenjiangChina
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro‐Psycho‐Diseases, College of Pharmaceutical ScienceSoochow UniversitySuzhouChina
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Chunhong Jiang
- State Key Laboratory of Innovative Nature Medicine and TCM InjectionsJiangxi Qingfeng Pharmaceutical Co., Ltd.GanzhouChina
| | - Ting Fan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Zheng‐Hong Qin
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro‐Psycho‐Diseases, College of Pharmaceutical ScienceSoochow UniversitySuzhouChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life SciencesNanjing UniversityNanjingChina
| | - Jing Gao
- School of PharmacyJiangsu UniversityZhenjiangChina
| |
Collapse
|
22
|
|
23
|
Theme 7 Pre-clinical therapeutic strategies. Amyotroph Lateral Scler Frontotemporal Degener 2018; 19:217-239. [DOI: 10.1080/21678421.2018.1510574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
24
|
MacDougall G, Anderton RS, Mastaglia FL, Knuckey NW, Meloni BP. Mitochondria and neuroprotection in stroke: Cationic arginine-rich peptides (CARPs) as a novel class of mitochondria-targeted neuroprotective therapeutics. Neurobiol Dis 2018; 121:17-33. [PMID: 30218759 DOI: 10.1016/j.nbd.2018.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/26/2018] [Accepted: 09/11/2018] [Indexed: 01/11/2023] Open
Abstract
Stroke is the second leading cause of death globally and represents a major cause of devastating long-term disability. Despite sustained efforts to develop clinically effective neuroprotective therapies, presently there is no clinically available neuroprotective agent for stroke. As a central mediator of neurodamaging events in stroke, mitochondria are recognised as a critical neuroprotective target, and as such, provide a focus for developing mitochondrial-targeted therapeutics. In recent years, cationic arginine-rich peptides (CARPs) have been identified as a novel class of neuroprotective agent with several demonstrated mechanisms of action, including their ability to target mitochondria and exert positive effects on the organelle. This review provides an overview on neuronal mitochondrial dysfunction in ischaemic stroke pathophysiology and highlights the potential beneficial effects of CARPs on mitochondria in the ischaemic brain following stroke.
Collapse
Affiliation(s)
- Gabriella MacDougall
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; School of Heath Sciences, and Institute for Health Research, The University Notre Dame Australia, Fremantle, Australia.
| | - Ryan S Anderton
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; School of Heath Sciences, and Institute for Health Research, The University Notre Dame Australia, Fremantle, Australia
| | - Frank L Mastaglia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Neville W Knuckey
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Bruno P Meloni
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Perron Institute for Neurological and Translational Science, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| |
Collapse
|
25
|
Edwards AB, Anderton RS, Knuckey NW, Meloni BP. Assessment of therapeutic window for poly-arginine-18D (R18D) in a P7 rat model of perinatal hypoxic-ischaemic encephalopathy. J Neurosci Res 2018; 96:1816-1826. [DOI: 10.1002/jnr.24315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Adam B. Edwards
- Perron Institute for Neurological and Translational Science, QEII Medical Centre; Nedlands Western Australia Australia
- School of Health Sciences and Institute for Health Research; The University of Notre Dame Australia; Fremantle Western Australia Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital; QEII Medical Centre; Nedlands Western Australia Australia
| | - Ryan S. Anderton
- Perron Institute for Neurological and Translational Science, QEII Medical Centre; Nedlands Western Australia Australia
- School of Health Sciences and Institute for Health Research; The University of Notre Dame Australia; Fremantle Western Australia Australia
- Centre for Neuromuscular and Neurological Disorders; The University of Western Australia; Nedlands Western Australia Australia
| | - Neville W. Knuckey
- Perron Institute for Neurological and Translational Science, QEII Medical Centre; Nedlands Western Australia Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital; QEII Medical Centre; Nedlands Western Australia Australia
- Centre for Neuromuscular and Neurological Disorders; The University of Western Australia; Nedlands Western Australia Australia
| | - Bruno P. Meloni
- Perron Institute for Neurological and Translational Science, QEII Medical Centre; Nedlands Western Australia Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital; QEII Medical Centre; Nedlands Western Australia Australia
- Centre for Neuromuscular and Neurological Disorders; The University of Western Australia; Nedlands Western Australia Australia
| |
Collapse
|
26
|
Perinatal Hypoxic-Ischemic Encephalopathy and Neuroprotective Peptide Therapies: A Case for Cationic Arginine-Rich Peptides (CARPs). Brain Sci 2018; 8:brainsci8080147. [PMID: 30087289 PMCID: PMC6119922 DOI: 10.3390/brainsci8080147] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) is the leading cause of mortality and morbidity in neonates, with survivors suffering significant neurological sequelae including cerebral palsy, epilepsy, intellectual disability and autism spectrum disorders. While hypothermia is used clinically to reduce neurological injury following HIE, it is only used for term infants (>36 weeks gestation) in tertiary hospitals and improves outcomes in only 30% of patients. For these reasons, a more effective and easily administrable pharmacological therapeutic agent, that can be used in combination with hypothermia or alone when hypothermia cannot be applied, is urgently needed to treat pre-term (≤36 weeks gestation) and term infants suffering HIE. Several recent studies have demonstrated that cationic arginine-rich peptides (CARPs), which include many cell-penetrating peptides [CPPs; e.g., transactivator of transcription (TAT) and poly-arginine-9 (R9; 9-mer of arginine)], possess intrinsic neuroprotective properties. For example, we have demonstrated that poly-arginine-18 (R18; 18-mer of arginine) and its D-enantiomer (R18D) are neuroprotective in vitro following neuronal excitotoxicity, and in vivo following perinatal hypoxia-ischemia (HI). In this paper, we review studies that have used CARPs and other peptides, including putative neuroprotective peptides fused to TAT, in animal models of perinatal HIE. We critically evaluate the evidence that supports our hypothesis that CARP neuroprotection is mediated by peptide arginine content and positive charge and that CARPs represent a novel potential therapeutic for HIE.
Collapse
|
27
|
A lincRNA-p21/miR-181 family feedback loop regulates microglial activation during systemic LPS- and MPTP- induced neuroinflammation. Cell Death Dis 2018; 9:803. [PMID: 30038357 PMCID: PMC6056543 DOI: 10.1038/s41419-018-0821-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/12/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022]
Abstract
The role of microglial-mediated sustained neuroinflammation in the onset and progression of Parkinson’s disease (PD) is well established, but the mechanisms contributing to microglial activation remain unclear. LincRNA-p21, a well studied long intergenic noncoding RNA (lincRNA), plays pivotal roles in diverse biological processes and diseases. Its role in microglial activation and inflammation-induced neurotoxicity, however, has not yet been fully elucidated. Here, we report that lincRNA-p21 promotes microglial activation through a p53-dependent transcriptional pathway. We further demonstrate that lincRNA-p21 competitively binds to the miR-181 family and induces microglial activation through the miR-181/PKC-δ pathway. Moreover, PKC-δ induction further increases the expression of p53/lincRNA-p21 and thus forms a circuit. Taken together, our results suggest that p53/lincRNA-p21, together with miR-181/PKC-δ, form a double-negative feedback loop that facilitates sustained microglial activation and the deterioration of neurodegeneration.
Collapse
|
28
|
Szelechowski M, Amoedo N, Obre E, Léger C, Allard L, Bonneu M, Claverol S, Lacombe D, Oliet S, Chevallier S, Le Masson G, Rossignol R. Metabolic Reprogramming in Amyotrophic Lateral Sclerosis. Sci Rep 2018; 8:3953. [PMID: 29500423 PMCID: PMC5834494 DOI: 10.1038/s41598-018-22318-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction in the spinal cord is a hallmark of amyotrophic lateral sclerosis (ALS), but the neurometabolic alterations during early stages of the disease remain unknown. Here, we investigated the bioenergetic and proteomic changes in ALS mouse motor neurons and patients' skin fibroblasts. We first observed that SODG93A mice presymptomatic motor neurons display alterations in the coupling efficiency of oxidative phosphorylation, along with fragmentation of the mitochondrial network. The proteome of presymptomatic ALS mice motor neurons also revealed a peculiar metabolic signature with upregulation of most energy-transducing enzymes, including the fatty acid oxidation (FAO) and the ketogenic components HADHA and ACAT2, respectively. Accordingly, FAO inhibition altered cell viability specifically in ALS mice motor neurons, while uncoupling protein 2 (UCP2) inhibition recovered cellular ATP levels and mitochondrial network morphology. These findings suggest a novel hypothesis of ALS bioenergetics linking FAO and UCP2. Lastly, we provide a unique set of data comparing the molecular alterations found in human ALS patients' skin fibroblasts and SODG93A mouse motor neurons, revealing conserved changes in protein translation, folding and assembly, tRNA aminoacylation and cell adhesion processes.
Collapse
Affiliation(s)
- M Szelechowski
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - N Amoedo
- Bordeaux University, 33000, Bordeaux, France
- INSERM U1211, MRGM, 33000, Bordeaux, France
| | - E Obre
- CELLOMET, Center of Functional Genomics (CGFB), 146 Rue Léo Saignat, 33000, Bordeaux, France
| | - C Léger
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - L Allard
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - M Bonneu
- Bordeaux University, 33000, Bordeaux, France
- Center of Functional Genomics (CGFB), Proteomic Facility, Bordeaux University, 33000, Bordeaux, France
| | - S Claverol
- Bordeaux University, 33000, Bordeaux, France
- Center of Functional Genomics (CGFB), Proteomic Facility, Bordeaux University, 33000, Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000, Bordeaux, France
- INSERM U1211, MRGM, 33000, Bordeaux, France
| | - S Oliet
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - S Chevallier
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - G Le Masson
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France.
- Bordeaux University, 33000, Bordeaux, France.
| | - R Rossignol
- Bordeaux University, 33000, Bordeaux, France.
- INSERM U1211, MRGM, 33000, Bordeaux, France.
| |
Collapse
|
29
|
Poly-arginine R18 and R18D (D-enantiomer) peptides reduce infarct volume and improves behavioural outcomes following perinatal hypoxic-ischaemic encephalopathy in the P7 rat. Mol Brain 2018; 11:8. [PMID: 29426351 PMCID: PMC5810179 DOI: 10.1186/s13041-018-0352-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
We examined the neuroprotective efficacy of the poly-arginine peptide R18 and its D-enantiomer R18D in a perinatal hypoxic-ischaemic (HI) model in P7 Sprague-Dawley rats. R18 and R18D peptides were administered intraperitoneally at doses of 30, 100, 300 or 1000 nmol/kg immediately after HI (8% O2/92%N2 for 2.5 h). The previously characterised neuroprotective JNKI-1-TATD peptide at a dose of 1000 nmol/kg was used as a control. Infarct volume and behavioural outcomes were measured 48 h after HI. For the R18 and R18D doses examined, total infarct volume was reduced by 25.93% to 43.80% (P = 0.038 to < 0.001). By comparison, the JNKI-1-TATD reduced lesion volume by 25.27% (P = 0.073). Moreover, R18 and R18D treatment resulted in significant improvements in behavioural outcomes, while with JNKI-1-TATD there was a trend towards improvement. As an insight into the likely mechanism underlying the effects of R18, R18D and JNKI-1-TATD, the peptides were added to cortical neuronal cultures exposed to glutamic acid excitotoxicity, resulting in up to 89, 100 and 71% neuroprotection, respectively, and a dose dependent inhibition of neuronal calcium influx. The study further confirms the neuroprotective properties of poly-arginine peptides, and suggests a potential therapeutic role for R18 and R18D in the treatment of HIE.
Collapse
|
30
|
Hippocampal expression of a virus-derived protein impairs memory in mice. Proc Natl Acad Sci U S A 2018; 115:1611-1616. [PMID: 29378968 DOI: 10.1073/pnas.1711977115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The analysis of the biology of neurotropic viruses, notably of their interference with cellular signaling, provides a useful tool to get further insight into the role of specific pathways in the control of behavioral functions. Here, we exploited the natural property of a viral protein identified as a major effector of behavioral disorders during infection. We used the phosphoprotein (P) of Borna disease virus, which acts as a decoy substrate for protein kinase C (PKC) when expressed in neurons and disrupts synaptic plasticity. By a lentiviral-based strategy, we directed the singled-out expression of P in the dentate gyrus of the hippocampus and we examined its impact on mouse behavior. Mice expressing the P protein displayed increased anxiety and impaired long-term memory in contextual and spatial memory tasks. Interestingly, these effects were dependent on P protein phosphorylation by PKC, as expression of a mutant form of P devoid of its PKC phosphorylation sites had no effect on these behaviors. We also revealed features of behavioral impairment induced by P protein expression but that were independent of its phosphorylation by PKC. Altogether, our findings provide insight into the behavioral correlates of viral infection, as well as into the impact of virus-mediated alterations of the PKC pathway on behavioral functions.
Collapse
|
31
|
Tan VX, Lassus B, Lim CK, Tixador P, Courte J, Bessede A, Guillemin GJ, Peyrin JM. Neurotoxicity of the Cyanotoxin BMAA Through Axonal Degeneration and Intercellular Spreading. Neurotox Res 2017; 33:62-75. [PMID: 28842862 DOI: 10.1007/s12640-017-9790-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/20/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022]
Abstract
β-Methylamino-L-alanine (BMAA) is implicated in neurodegeneration and neurotoxicity, particularly in ALS-Parkinson Dementia Complex. Neurotoxic properties of BMAA have been partly elucidated, while its transcellular spreading capacity has not been examined. Using reconstructed neuronal networks in microfluidic chips, separating neuronal cells into two subcompartments-(1) the proximal, containing first-order neuronal soma and dendrites, and (2) a distal compartment, containing either only axons originating from first-order neurons or second-order striatal neurons-creates a cortico-striatal network. Using this system, we investigated the toxicity and spreading of BMAA in murine primary neurons. We used a newly developed antibody to detect BMAA in cells. After treatment with 10 μM BMAA, the cyanotoxin was incorporated in first-degree neurons. We also observed a rapid trans-neuronal spread of BMAA to unexposed second-degree neurons in 48 h, followed by axonal degeneration, with limited somatic death. This in vitro study demonstrates BMAA axonal toxicity at sublethal concentrations and, for the first time, the transcellular spreading abilities of BMAA. This neuronal dying forward spread that could possibly be associated with progression of some neurodegenerative diseases especially amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Vanessa X Tan
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | - Benjamin Lassus
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | - Chai K Lim
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Philippe Tixador
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | - Josquin Courte
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France
| | | | - Gilles J Guillemin
- Macquarie University Centre for MND Research, Department of Biological Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Jean-Michel Peyrin
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine, Adaptation Biologique et vieillissement, F-75005, Paris, France.
| |
Collapse
|
32
|
Das B, Rajagopalan S, Joshi GS, Xu L, Luo D, Andersen JK, Todi SV, Dutta AK. A novel iron (II) preferring dopamine agonist chelator D-607 significantly suppresses α-syn- and MPTP-induced toxicities in vivo. Neuropharmacology 2017; 123:88-99. [PMID: 28533164 DOI: 10.1016/j.neuropharm.2017.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 12/28/2022]
Abstract
Here, we report the characterization of a novel hybrid D2/D3 agonist and iron (II) specific chelator, D-607, as a multi-target-directed ligand against Parkinson's disease (PD). In our previously published report, we showed that D-607 is a potent agonist of dopamine (DA) D2/D3 receptors, exhibits efficacy in a reserpinized PD animal model and preferentially chelates to iron (II). As further evidence of its potential as a neuroprotective agent in PD, the present study reveals D-607 to be protective in neuronal PC12 cells against 6-OHDA toxicity. In an in vivo Drosophila melanogaster model expressing a disease-causing variant of α-synuclein (α-Syn) protein in fly eyes, the compound was found to significantly suppress toxicity compared to controls, concomitant with reduced levels of aggregated α-Syn. Furthermore, D-607 was able to rescue DAergic neurons from MPTP toxicity in mice, a well-known PD neurotoxicity model, following both sub-chronic and chronic MPTP administration. Mechanistic studies indicated that possible protection of mitochondria, up-regulation of hypoxia-inducible factor, reduction in formation of α-Syn aggregates and antioxidant activity may underlie the observed neuroprotection effects. These observations strongly suggest that D-607 has potential as a promising multifunctional lead molecule for viable symptomatic and disease-modifying therapy for PD.
Collapse
Affiliation(s)
- Banibrata Das
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA
| | | | - Gnanada S Joshi
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Liping Xu
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Dan Luo
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Julie K Andersen
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945, USA
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Aloke K Dutta
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
33
|
MacDougall G, Anderton RS, Edwards AB, Knuckey NW, Meloni BP. The Neuroprotective Peptide Poly-Arginine-12 (R12) Reduces Cell Surface Levels of NMDA NR2B Receptor Subunit in Cortical Neurons; Investigation into the Involvement of Endocytic Mechanisms. J Mol Neurosci 2016; 61:235-246. [PMID: 27866326 DOI: 10.1007/s12031-016-0861-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/11/2016] [Indexed: 10/20/2022]
Abstract
We have previously reported that cationic poly-arginine and arginine-rich cell-penetrating peptides display high-level neuroprotection and reduce calcium influx following in vitro excitotoxicity, as well as reduce brain injury in animal stroke models. Using the neuroprotective peptides poly-arginine R12 (R12) and the NR2B9c peptide fused to the arginine-rich carrier peptide TAT (TAT-NR2B9c; also known as NA-1), we investigated the mechanisms whereby poly-arginine and arginine-rich peptides reduce glutamate-induced excitotoxic calcium influx. Using cell surface biotin protein labeling and western blot analysis, we demonstrated that R12 and TAT-NR2B9c significantly reduced cortical neuronal cell surface expression of the NMDA receptor subunit NR2B. Chemical endocytic inhibitors used individually or in combination prior to glutamate excitotoxicity did not significantly affect R12 peptide neuroprotective efficacy. Similarly, pretreatment of neurons with enzymes to degrade anionic cell surface proteoglycans, heparan sulfate proteoglycan (HSPG), and chondroitin sulfate proteoglycan (CSPG), as well as sialic acid residues, did not significantly affect peptide neuroprotective efficacy. While the exact mechanisms responsible for R12 peptide-mediated NMDA receptor NR2B subunit cell surface downregulation were not identified, an endocytic process could not be ruled out. The study supports our hypothesis that arginine-rich peptides reduce excitotoxic calcium influx by reducing the levels of cell surface ion channels.
Collapse
Affiliation(s)
- Gabriella MacDougall
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA, Australia. .,Western Australian Neuroscience Research Institute, A Block, 4th Floor, QEII Medical Centre, Verdun St, Nedlands, WA, 6009, Australia. .,School of Heath Sciences, The University Notre Dame, Fremantle, WA, Australia.
| | - Ryan S Anderton
- Western Australian Neuroscience Research Institute, A Block, 4th Floor, QEII Medical Centre, Verdun St, Nedlands, WA, 6009, Australia.,School of Heath Sciences, The University Notre Dame, Fremantle, WA, Australia
| | - Adam B Edwards
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA, Australia.,Western Australian Neuroscience Research Institute, A Block, 4th Floor, QEII Medical Centre, Verdun St, Nedlands, WA, 6009, Australia.,School of Heath Sciences, The University Notre Dame, Fremantle, WA, Australia
| | - Neville W Knuckey
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA, Australia.,Western Australian Neuroscience Research Institute, A Block, 4th Floor, QEII Medical Centre, Verdun St, Nedlands, WA, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia
| | - Bruno P Meloni
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA, Australia.,Western Australian Neuroscience Research Institute, A Block, 4th Floor, QEII Medical Centre, Verdun St, Nedlands, WA, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia
| |
Collapse
|
34
|
Grehl S, Martina D, Goyenvalle C, Deng ZD, Rodger J, Sherrard RM. In vitro Magnetic Stimulation: A Simple Stimulation Device to Deliver Defined Low Intensity Electromagnetic Fields. Front Neural Circuits 2016; 10:85. [PMID: 27857683 PMCID: PMC5093126 DOI: 10.3389/fncir.2016.00085] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/10/2016] [Indexed: 01/10/2023] Open
Abstract
Non-invasive brain stimulation (NIBS) by electromagnetic fields appears to benefit human neurological and psychiatric conditions, although the optimal stimulation parameters and underlying mechanisms remain unclear. Although, in vitro studies have begun to elucidate cellular mechanisms, stimulation is delivered by a range of coils (from commercially available human stimulation coils to laboratory-built circuits) so that the electromagnetic fields induced within the tissue to produce the reported effects are ill-defined. Here, we develop a simple in vitro stimulation device with plug-and-play features that allow delivery of a range of stimulation parameters. We chose to test low intensity repetitive magnetic stimulation (LI-rMS) delivered at three frequencies to hindbrain explant cultures containing the olivocerebellar pathway. We used computational modeling to define the parameters of a stimulation circuit and coil that deliver a unidirectional homogeneous magnetic field of known intensity and direction, and therefore a predictable electric field, to the target. We built the coil to be compatible with culture requirements: stimulation within an incubator; a flat surface allowing consistent position and magnetic field direction; location outside the culture plate to maintain sterility and no heating or vibration. Measurements at the explant confirmed the induced magnetic field was homogenous and matched the simulation results. To validate our system we investigated biological effects following LI-rMS at 1 Hz, 10 Hz and biomimetic high frequency, which we have previously shown induces neural circuit reorganization. We found that gene expression was modified by LI-rMS in a frequency-related manner. Four hours after a single 10-min stimulation session, the number of c-fos positive cells increased, indicating that our stimulation activated the tissue. Also, after 14 days of LI-rMS, the expression of genes normally present in the tissue was differentially modified according to the stimulation delivered. Thus we describe a simple magnetic stimulation device that delivers defined stimulation parameters to different neural systems in vitro. Such devices are essential to further understanding of the fundamental effects of magnetic stimulation on biological tissue and optimize therapeutic application of human NIBS.
Collapse
Affiliation(s)
- Stephanie Grehl
- Sorbonne Universités, UPMC Univ Paris 06 & CNRS, IBPS-B2A, UMR 8256 Biological Adaptation and AgeingParis, France; Experimental and Regenerative Neuroscience, School of Animal Biology, the University of Western Australia, PerthWA, Australia
| | - David Martina
- Institut Langevin, ESPCI ParisTech & CNRS, UMR7587 INSERM ERL U979 Paris, France
| | - Catherine Goyenvalle
- Sorbonne Universités, UPMC Univ Paris 06 & CNRS, IBPS-B2A, UMR 8256 Biological Adaptation and Ageing Paris, France
| | - Zhi-De Deng
- Non-invasive Neuromodulation Unit, Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, BethesdaMD, USA; Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, DurhamNC, USA
| | - Jennifer Rodger
- Experimental and Regenerative Neuroscience, School of Animal Biology, the University of Western Australia, Perth WA, Australia
| | - Rachel M Sherrard
- Sorbonne Universités, UPMC Univ Paris 06 & CNRS, IBPS-B2A, UMR 8256 Biological Adaptation and Ageing Paris, France
| |
Collapse
|
35
|
Lassus B, Magnifico S, Pignon S, Belenguer P, Miquel MC, Peyrin JM. Alterations of mitochondrial dynamics allow retrograde propagation of locally initiated axonal insults. Sci Rep 2016; 6:32777. [PMID: 27604820 PMCID: PMC5015069 DOI: 10.1038/srep32777] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/08/2016] [Indexed: 12/15/2022] Open
Abstract
In chronic neurodegenerative syndromes, neurons progressively die through a generalized retraction pattern triggering retrograde axonal degeneration toward the cell bodies, which molecular mechanisms remain elusive. Recent observations suggest that direct activation of pro-apoptotic signaling in axons triggers local degenerative events associated with early alteration of axonal mitochondrial dynamics. This raises the question of the role of mitochondrial dynamics on both axonal vulnerability stress and their implication in the spreading of damages toward unchallenged parts of the neuron. Here, using microfluidic chambers, we assessed the consequences of interfering with OPA1 and DRP1 proteins on axonal degeneration induced by local application of rotenone. We found that pharmacological inhibition of mitochondrial fission prevented axonal damage induced by rotenone, in low glucose conditions. While alteration of mitochondrial dynamics per se did not lead to spontaneous axonal degeneration, it dramatically enhanced axonal vulnerability to rotenone, which had no effect in normal glucose conditions, and promoted retrograde spreading of axonal degeneration toward the cell body. Altogether, our results suggest a mitochondrial priming effect in axons as a key process of axonal degeneration. In the context of neurodegenerative diseases, like Parkinson's and Alzheimer's, mitochondria fragmentation could hasten neuronal death and initiate spatial dispersion of locally induced degenerative events.
Collapse
Affiliation(s)
- Benjamin Lassus
- CNRS UMR 8256, Biological Adaptation and Ageing, Paris, 75005, France.,Sorbonne Universités, UPMC, Institut de Biologie Paris-Seine, Paris, 75005, France
| | - Sebastien Magnifico
- CNRS UMR 8256, Biological Adaptation and Ageing, Paris, 75005, France.,Sorbonne Universités, UPMC, Institut de Biologie Paris-Seine, Paris, 75005, France
| | - Sandra Pignon
- CNRS UMR 8256, Biological Adaptation and Ageing, Paris, 75005, France.,Sorbonne Universités, UPMC, Institut de Biologie Paris-Seine, Paris, 75005, France
| | - Pascale Belenguer
- CNRS UMR 5169 Research Center on Animal Cognition, Center for Integrative Biology, Toulouse University, Université Toulouse 3 Paul Sabatier, 31400, France
| | - Marie-Christine Miquel
- CNRS UMR 5169 Research Center on Animal Cognition, Center for Integrative Biology, Toulouse University, Université Toulouse 3 Paul Sabatier, 31400, France
| | - Jean-Michel Peyrin
- CNRS UMR 8256, Biological Adaptation and Ageing, Paris, 75005, France.,Sorbonne Universités, UPMC, Institut de Biologie Paris-Seine, Paris, 75005, France
| |
Collapse
|
36
|
The Cytomegalovirus protein pUL37×1 targets mitochondria to mediate neuroprotection. Sci Rep 2016; 6:31373. [PMID: 27562039 PMCID: PMC4999870 DOI: 10.1038/srep31373] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
There is substantial evidence that mitochondrial dysfunction plays a significant role in the pathogenesis of Parkinson disease (PD). This contribution probably encompasses defects of oxidative phosphorylation, mitochondrial turnover (mitophagy), mitochondrial derived oxidative stress, and apoptotic signalling. Human cytomegalovirus immediate-early protein pUL37 × 1 induces Bax mitochondrial translocation and inactivation to prevent apoptosis. Over-expressing pUL37 × 1 in neuronal cells protects against staurosporin and 6-hydroxydopamine induced apoptosis and cell death. Protection is not enhanced by bax silencing in pUL37 × 1 over-expressing cells, suggesting a bax-dependent mechanism of action. pUL37 × 1 increases glycolysis and induces mitochondrial hyperpolarization, a bax independent anti-apoptotic action. pUL37 × 1 increases glycolysis through activation of phosphofructokinase by a calcium-dependent pathway. The dual anti-apoptotic mechanism of pUL37 × 1 may be considered a novel neuroprotective strategy in diseases where mitochondrial dysfunction and apoptotic pathways are involved.
Collapse
|
37
|
Endres K, Reinhardt S, Geladaris A, Knies J, Grimm M, Hartmann T, Schmitt U. Transnasal delivery of human A-beta peptides elicits impaired learning and memory performance in wild type mice. BMC Neurosci 2016; 17:44. [PMID: 27377996 PMCID: PMC4932715 DOI: 10.1186/s12868-016-0280-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/24/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Murine models of Alzheimer's disease (AD) are mainly based on overexpression of pathologic amyloid precursor protein and/or presenilins. Those genes resemble underlying cause of early onset type of AD while about 99 % of all human cases are to be characterized as sporadic, late onset. Appropriate animal models for this type of AD are still missing. We here investigated, if transnasal delivery of A-beta 42 peptides might serve to mimic pathological effects in mice. RESULTS A-beta 42 peptides, used for the behavioral study, showed the expected dose-dependent toxicity in neur oblastoma cell line SH-SY5Y and were able to form higher molecular weight species in vitro. Upon delivery into nostrils of wild type mice, protein bands that might represent aggregation products of the exogenously applied human A-beta 42 were only observed in total brain homogenates from mice pre-treated with mannitol. By using TAMRA-labeled A-beta 42 peptides we demonstrated, that transport throughout the brain was achieved already 1 h after administration. FVB/N mice treated with A-beta 42 for 3 days were significantly impaired in the cue-retention condition of the fear conditioning task as compared to controls whereas A-beta-treated C57B6/J mice were impaired in the context condition. In the Morris water maze test, these mice also displayed a delayed learning performance, indicated by significantly longer time to find the platform. Those deficits were also seen for memory performance in the probe trial as measured by number of crossings of the former platform position and time spent in the goal quadrant. CONCLUSIONS Existing AD mouse models are of genetic origin and need prolonged housing time before onset of pathology. Our short-term treatment induced learning and memory deficits via exogenous application of A-beta peptides comparable to those observed for the transgenic animals. With the transnasal A-beta 42 treatment we present an approach to investigate purely A-beta related changes suitable as a model for symptoms of Alzheimer's dementia (AD). Resulting behavioral deficits were indicative for familial type of Alzheimer's disease as well as for the late onset variant.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany.
| | - Sven Reinhardt
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Anastasia Geladaris
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Julia Knies
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Marcus Grimm
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany.,Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Tobias Hartmann
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany.,Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Ulrich Schmitt
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| |
Collapse
|
38
|
Wang H, Liu J, Gao G, Wu X, Wang X, Yang H. Protection effect of piperine and piperlonguminine from Piper longum L. alkaloids against rotenone-induced neuronal injury. Brain Res 2016; 1639:214-27. [PMID: 26232071 DOI: 10.1016/j.brainres.2015.07.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 07/18/2015] [Accepted: 07/20/2015] [Indexed: 12/22/2022]
Abstract
Currently available treatment approaches for Parkinson׳s disease (PD) are limited in terms of variety and efficacy. Piper longum L. (PLL; Piperaceae) is used in traditional medicine in Asia and the Pacific Islands, with demonstrated anti-inflammatory and antioxidant activities in preclinical studies, and alkaloid extracts of PLL have shown protective effects in PD models. The present study investigated the mechanistic basis for the observed protective effects of PLL. Rats treated with PLL-derived alkaloids showed improvement in rotenone-induced motor deficits, while reactive oxygen species (ROS) production was decreased, mitochondrial membrane potential was stabilized, and the opening of the mitochondrial permeability transition pore (mPTP)-which is involved in ROS production-was inhibited. In addition, rotenone-induced apoptosis was abrogated in the presence of these alkaloids, while a pretreatment stimulated autophagy, likely mitigating neuronal injury by the removal of damaged mitochondria. These findings provide novel insight into the neuroprotective function of PLL as well as evidence in favor of its use in PD treatment. This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
Affiliation(s)
- Hao Wang
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Jia Liu
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Ge Gao
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Xia Wu
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Xiaomin Wang
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China
| | - Hui Yang
- Center of Parkinson׳s Disease Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Department of Neurobiology Capital Medical University, Beijing 100069, China.
| |
Collapse
|
39
|
Milani D, Knuckey NW, Anderton RS, Cross JL, Meloni BP. The R18 Polyarginine Peptide Is More Effective Than the TAT-NR2B9c (NA-1) Peptide When Administered 60 Minutes after Permanent Middle Cerebral Artery Occlusion in the Rat. Stroke Res Treat 2016; 2016:2372710. [PMID: 27247825 PMCID: PMC4877491 DOI: 10.1155/2016/2372710] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/11/2016] [Indexed: 11/24/2022] Open
Abstract
We examined the dose responsiveness of polyarginine R18 (100, 300, and 1000 nmol/kg) when administered 60 minutes after permanent middle cerebral artery occlusion (MCAO). The TAT-NR2B9c peptide, which is known to be neuroprotective in rodent and nonhuman primate stroke models, served as a positive control. At 24 hours after MCAO, there was reduced total infarct volume in R18 treated animals at all doses, but this reduction only reached statistical significance at doses of 100 and 1000 nmol/kg. The TAT-NR2B9c peptide reduced infarct volume at doses of 300 and 1000 nmol/kg, but not to a statistically significant extent, while the 100 nmol/kg dose was ineffective. The reduction in infarct volume with R18 and TAT-NR2B9c peptide treatments was mirrored by improvements in one or more functional outcomes (namely, neurological score, adhesive tape removal, and rota-rod), but not to a statistically significant extent. These findings further confirm the neuroprotective properties of polyarginine peptides and for R18 extend its therapeutic time window and dose range, as well as demonstrating its greater efficacy compared to TAT-NR2B9c in a severe stroke model. The superior neuroprotective efficacy of R18 over TAT-NR2B9c highlights the potential of this polyarginine peptide as a lead candidate for studies in human stroke.
Collapse
Affiliation(s)
- D. Milani
- School of Health Sciences, The University of Notre Dame Australia, Fremantle, WA 6160, Australia
- Western Australian Neuroscience Research Institute, Nedlands, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - N. W. Knuckey
- Western Australian Neuroscience Research Institute, Nedlands, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| | - R. S. Anderton
- School of Health Sciences, The University of Notre Dame Australia, Fremantle, WA 6160, Australia
- Western Australian Neuroscience Research Institute, Nedlands, WA 6009, Australia
| | - J. L. Cross
- Western Australian Neuroscience Research Institute, Nedlands, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| | - B. P. Meloni
- Western Australian Neuroscience Research Institute, Nedlands, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA 6009, Australia
| |
Collapse
|
40
|
Sun Q, Weinger JG, Mao F, Liu G. Regulation of structural and functional synapse density by L-threonate through modulation of intraneuronal magnesium concentration. Neuropharmacology 2016; 108:426-39. [PMID: 27178134 DOI: 10.1016/j.neuropharm.2016.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/18/2016] [Accepted: 05/09/2016] [Indexed: 12/25/2022]
Abstract
Oral administration of the combination of L-threonate (threonate) and magnesium (Mg(2+)) in the form of L-Threonic acid Magnesium salt (L-TAMS) can enhance learning and memory in young rats and prevent memory decline in aging rats and in Alzheimer's disease model mice. Recent results from a human clinical trial demonstrate the efficacy of L-TAMS in restoring global cognitive abilities of older adults. Previously, we reported that neuronal intracellular Mg(2+) serves as a critical signaling molecule for controlling synapse density, a key factor that determines cognitive ability. The elevation of brain Mg(2+) by oral administration of L-TAMS in intact animals plays a significant role in mediating the therapeutic effects of L-TAMS. The current study sought to elucidate the unique role of threonate. We aimed to understand if threonate acts directly to elevate intraneuronal Mg(2+), and why Mg(2+) given without threonate is ineffective for enhancing learning and memory ability. We discovered that threonate is naturally present in cerebrospinal fluid (CSF) and oral treatment with L-TAMS elevated CSF threonate. In cultured hippocampal neurons, threonate treatment directly induced an increase in intracellular Mg(2+) concentration. Functionally, elevating threonate upregulated expression of NR2B-containing NMDAR, boosted mitochondrial membrane potential (ΔΨm), and increased functional synapse density in neuronal cultures. These effects are unique to threonate, as other common Mg(2+) anions failed to have the same results. Mechanistically, threonate's effects were specifically mediated through glucose transporters (GLUTs). We also evaluated the effects of threonate in human neural stem cell-derived neurons, and found it was equally effective at upregulating synapse density. The current study provides an explanation for why threonate is an essential component of L-TAMS and supports the use of L-TAMS to promote cognitive abilities in human.
Collapse
Affiliation(s)
- Qifeng Sun
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | | | - Fei Mao
- Neurocentria, Inc., Fremont, CA 94538, USA
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China; Neurocentria, Inc., Fremont, CA 94538, USA.
| |
Collapse
|
41
|
|
42
|
Ferré CA, Davezac N, Thouard A, Peyrin JM, Belenguer P, Miquel MC, Gonzalez-Dunia D, Szelechowski M. Manipulation of the N-terminal sequence of the Borna disease virus X protein improves its mitochondrial targeting and neuroprotective potential. FASEB J 2015; 30:1523-33. [PMID: 26700735 DOI: 10.1096/fj.15-279620] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/08/2015] [Indexed: 01/11/2023]
Abstract
To favor their replication, viruses express proteins that target diverse mammalian cellular pathways. Due to the limited size of many viral genomes, such proteins are endowed with multiple functions, which require targeting to different subcellular compartments. One salient example is the X protein of Borna disease virus, which is expressed both at the mitochondria and in the nucleus. Moreover, we recently demonstrated that mitochondrial X protein is neuroprotective. In this study, we sought to examine the mechanisms whereby the X protein transits between subcellular compartments and to define its localization signals, to enhance its mitochondrial accumulation and thus, potentially, its neuroprotective activity. We transfected plasmids expressing fusion proteins bearing different domains of X fused to enhanced green fluorescent protein (eGFP) and compared their subcellular localization to that of eGFP. We observed that the 5-16 domain of X was responsible for both nuclear export and mitochondrial targeting and identified critical residues for mitochondrial localization. We next took advantage of these findings and constructed mutant X proteins that were targeted only to the mitochondria. Such mutants exhibited enhanced neuroprotective properties in compartmented cultures of neurons grown in microfluidic chambers, thereby confirming the parallel between mitochondrial accumulation of the X protein and its neuroprotective potential.-Ferré C. A., Davezac, N., Thouard, A., Peyrin, J. M., Belenguer, P., Miquel, M.-C., Gonzalez-Dunia, D., Szelechowski, M. Manipulation of the N-terminal sequence of the Borna disease virus X protein improves its mitochondrial targeting and neuroprotective potential.
Collapse
Affiliation(s)
- Cécile A Ferré
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Noélie Davezac
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Anne Thouard
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Jean-Michel Peyrin
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Pascale Belenguer
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Marie-Christine Miquel
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Daniel Gonzalez-Dunia
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Marion Szelechowski
- *INSERM, Unité Mixte de Recherche (UMR) 1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France; Centre National de la Recherche Scientifique (CNRS), UMR 5282, Toulouse, France; Université Toulouse III Paul Sabatier, Toulouse, France; CNRS UMR 5547, Centre de Biologie du Développement, Toulouse, France; CNRS UMR 8256, Biological Adaptation and Aging, Institut de Biologie Paris Seine, Université Pierre et Marie Curie, Paris, France; and Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| |
Collapse
|