1
|
Yu Y, Chen CZ, Celardo I, Tan BWZ, Hurcomb JD, Leal NS, Popovic R, Loh SHY, Martins LM. Enhancing mitochondrial one-carbon metabolism is neuroprotective in Alzheimer's disease models. Cell Death Dis 2024; 15:856. [PMID: 39582067 PMCID: PMC11586400 DOI: 10.1038/s41419-024-07179-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024]
Abstract
Alzheimer's disease (AD) is the most common form of age-related dementia. In AD, the death of neurons in the central nervous system is associated with the accumulation of toxic amyloid β peptide (Aβ) and mitochondrial dysfunction. Mitochondria are signal transducers of metabolic and biochemical information, and their impairment can compromise cellular function. Mitochondria compartmentalise several pathways, including folate-dependent one-carbon (1C) metabolism and electron transport by respiratory complexes. Mitochondrial 1C metabolism is linked to electron transport through complex I of the respiratory chain. Here, we analysed the proteomic changes in a fly model of AD by overexpressing a toxic form of Aβ (Aβ-Arc). We found that expressing Aβ-Arc caused alterations in components of both complex I and mitochondrial 1C metabolism. Genetically enhancing mitochondrial 1C metabolism through Nmdmc improved mitochondrial function and was neuroprotective in fly models of AD. We also found that exogenous supplementation with the 1C donor folinic acid improved mitochondrial health in both mammalian cells and fly models of AD. We found that genetic variations in MTHFD2L, the human orthologue of Nmdmc, were linked to AD risk. Additionally, Mendelian randomisation showed that increased folate intake decreased the risk of developing AD. Overall, our data suggest enhancement of folate-dependent 1C metabolism as a viable strategy to delay the progression and attenuate the severity of AD.
Collapse
Affiliation(s)
- Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK.
- Healthspan Biotics Ltd, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge, UK.
| | - Civia Z Chen
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Ivana Celardo
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Bryan Wei Zhi Tan
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - James D Hurcomb
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Nuno Santos Leal
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Rebeka Popovic
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Samantha H Y Loh
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - L Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK.
| |
Collapse
|
2
|
Marx N, Ritter N, Disse P, Seebohm G, Busch KB. Detailed analysis of Mdivi-1 effects on complex I and respiratory supercomplex assembly. Sci Rep 2024; 14:19673. [PMID: 39187541 PMCID: PMC11347648 DOI: 10.1038/s41598-024-69748-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/08/2024] [Indexed: 08/28/2024] Open
Abstract
Several human diseases, including cancer and neurodegeneration, are associated with excessive mitochondrial fragmentation. In this context, mitochondrial division inhibitor (Mdivi-1) has been tested as a therapeutic to block the fission-related protein dynamin-like protein-1 (Drp1). Recent studies suggest that Mdivi-1 interferes with mitochondrial bioenergetics and complex I function. Here we show that the molecular mechanism of Mdivi-1 is based on inhibition of complex I at the IQ site. This leads to the destabilization of complex I, impairs the assembly of N- and Q-respirasomes, and is associated with increased ROS production and reduced efficiency of ATP generation. Second, the calcium homeostasis of cells is impaired, which for example affects the electrical activity of neurons. Given the results presented here, a potential therapeutic application of Mdivi-1 is challenging because of its potential impact on synaptic activity. Similar to the Complex I inhibitor rotenone, Mdivi-1 may lead to neurodegenerative effects in the long term.
Collapse
Affiliation(s)
- Nico Marx
- Department of Biology, Institute of Integrative Cell Biology and Physiology (IIZP), University of Münster, Schloßplatz 5, 48149, Münster, Germany
| | - Nadine Ritter
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, 48149, Münster, Germany
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Paul Disse
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, 48149, Münster, Germany
| | - Guiscard Seebohm
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, 48149, Münster, Germany
| | - Karin B Busch
- Department of Biology, Institute of Integrative Cell Biology and Physiology (IIZP), University of Münster, Schloßplatz 5, 48149, Münster, Germany.
| |
Collapse
|
3
|
Venati SR, Uversky VN. Exploring Intrinsic Disorder in Human Synucleins and Associated Proteins. Int J Mol Sci 2024; 25:8399. [PMID: 39125972 PMCID: PMC11313516 DOI: 10.3390/ijms25158399] [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: 06/20/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In this work, we explored the intrinsic disorder status of the three members of the synuclein family of proteins-α-, β-, and γ-synucleins-and showed that although all three human synucleins are highly disordered, the highest levels of disorder are observed in γ-synuclein. Our analysis of the peculiarities of the amino acid sequences and modeled 3D structures of the human synuclein family members revealed that the pathological mutations A30P, E46K, H50Q, A53T, and A53E associated with the early onset of Parkinson's disease caused some increase in the local disorder propensity of human α-synuclein. A comparative sequence-based analysis of the synuclein proteins from various evolutionary distant species and evaluation of their levels of intrinsic disorder using a set of commonly used bioinformatics tools revealed that, irrespective of their origin, all members of the synuclein family analyzed in this study were predicted to be highly disordered proteins, indicating that their intrinsically disordered nature represents an evolutionary conserved and therefore functionally important feature. A detailed functional disorder analysis of the proteins in the interactomes of the human synuclein family members utilizing a set of commonly used disorder analysis tools showed that the human α-synuclein interactome has relatively higher levels of intrinsic disorder as compared with the interactomes of human β- and γ- synucleins and revealed that, relative to the β- and γ-synuclein interactomes, α-synuclein interactors are involved in a much broader spectrum of highly diversified functional pathways. Although proteins interacting with three human synucleins were characterized by highly diversified functionalities, this analysis also revealed that the interactors of three human synucleins were involved in three common functional pathways, such as the synaptic vesicle cycle, serotonergic synapse, and retrograde endocannabinoid signaling. Taken together, these observations highlight the critical importance of the intrinsic disorder of human synucleins and their interactors in various neuronal processes.
Collapse
Affiliation(s)
- Sriya Reddy Venati
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
4
|
Rodriguez P, Blakely RD. Sink or swim: Does a worm paralysis phenotype hold clues to neurodegenerative disease? J Cell Physiol 2024; 239:e31125. [PMID: 37795580 DOI: 10.1002/jcp.31125] [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: 07/15/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Receiving a neurodegenerative disease (NDD) diagnosis, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis, is devastating, particularly given the limited options for treatment. Advances in genetic technologies have allowed for efficient modeling of NDDs in animals and brought hope for new disease-modifying medications. The complexity of the mammalian brain and the costs and time needed to identify and develop therapeutic leads limits progress. Modeling NDDs in invertebrates, such as the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans, offers orders of magnitude increases in speed of genetic analysis and manipulation, and can be pursued at substantially reduced cost, providing an important, platform complement and inform research with mammalian NDD models. In this review, we describe how our efforts to exploit C. elegans for the study of neural signaling and health led to the discovery of a paralytic phenotype (swimming-induced paralysis) associated with altered dopamine signaling and, surprisingly, to the discovery of a novel gene and pathway whose dysfunction in glial cells triggers neurodegeneration. Research to date on swip-10 and its putative mammalian ortholog MBLAC1, suggests that a tandem analysis will offer insights into NDD mechanisms and insights into novel, disease-modifying therapeutics.
Collapse
Affiliation(s)
- Peter Rodriguez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Boca Raton, Florida, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Boca Raton, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
5
|
Wang X, Li S, Zhang C, Xu W, Wu M, Cheng J, Li Z, Tao L, Zhang Y. Stereoselective toxicity of acetochlor chiral isomers on the nervous system of zebrafish larvae. JOURNAL OF HAZARDOUS MATERIALS 2024; 464:133016. [PMID: 37992503 DOI: 10.1016/j.jhazmat.2023.133016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
Acetochlor (ACT) is a widely detected pesticide globally, and the neurotoxic effects of its chiral isomers on humans and environmental organisms remain uncertain. Zebrafish were used to study the neurotoxicity of ACT and its chiral isomers. Our study reveals that the R-ACT, Rac-ACT, and S-ACT induce neurotoxicity in zebrafish larvae by impairing vascular development and disrupting the blood-brain barrier. These detrimental effects lead to apoptosis in brain cells, hindered development of the central nervous system, and manifest as altered swimming behavior and social interactions in the larvae. Importantly, the neurotoxicity caused by the S-ACT exhibits the most pronounced impact and significantly diverges from the effects induced by the R-ACT. The neurotoxicity associated with the Rac-ACT falls intermediate between that of the R-ACT and S-ACT. Fascinatingly, we observed a remarkable recovery in the S-ACT-induced abnormalities in BBB, neurodevelopment, and behavior in zebrafish larvae upon supplementation of the Wnt/β-catenin signaling pathway. This observation strongly suggests that the Wnt/β-catenin signaling pathway serves as a major target of S-ACT-induced neurotoxicity in zebrafish larvae. In conclusion, S-ACT significantly influences zebrafish larval neurodevelopment by inhibiting the Wnt/β-catenin signaling pathway, distinguishing it from R-ACT neurotoxic effects.
Collapse
Affiliation(s)
- Xin Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Shoulin Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Zhang
- Department of Pathology, UT southwestern Medical Center, Dallas, TX 75390, United States
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Mengqi Wu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiagao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
6
|
Yap TA, Daver N, Mahendra M, Zhang J, Kamiya-Matsuoka C, Meric-Bernstam F, Kantarjian HM, Ravandi F, Collins ME, Francesco MED, Dumbrava EE, Fu S, Gao S, Gay JP, Gera S, Han J, Hong DS, Jabbour EJ, Ju Z, Karp DD, Lodi A, Molina JR, Baran N, Naing A, Ohanian M, Pant S, Pemmaraju N, Bose P, Piha-Paul SA, Rodon J, Salguero C, Sasaki K, Singh AK, Subbiah V, Tsimberidou AM, Xu QA, Yilmaz M, Zhang Q, Li Y, Bristow CA, Bhattacharjee MB, Tiziani S, Heffernan TP, Vellano CP, Jones P, Heijnen CJ, Kavelaars A, Marszalek JR, Konopleva M. Complex I inhibitor of oxidative phosphorylation in advanced solid tumors and acute myeloid leukemia: phase I trials. Nat Med 2023; 29:115-126. [PMID: 36658425 PMCID: PMC11975418 DOI: 10.1038/s41591-022-02103-8] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/21/2022] [Indexed: 01/21/2023]
Abstract
Although targeting oxidative phosphorylation (OXPHOS) is a rational anticancer strategy, clinical benefit with OXPHOS inhibitors has yet to be achieved. Here we advanced IACS-010759, a highly potent and selective small-molecule complex I inhibitor, into two dose-escalation phase I trials in patients with relapsed/refractory acute myeloid leukemia (NCT02882321, n = 17) and advanced solid tumors (NCT03291938, n = 23). The primary endpoints were safety, tolerability, maximum tolerated dose and recommended phase 2 dose (RP2D) of IACS-010759. The PK, PD, and preliminary antitumor activities of IACS-010759 in patients were also evaluated as secondary endpoints in both clinical trials. IACS-010759 had a narrow therapeutic index with emergent dose-limiting toxicities, including elevated blood lactate and neurotoxicity, which obstructed efforts to maintain target exposure. Consequently no RP2D was established, only modest target inhibition and limited antitumor activity were observed at tolerated doses, and both trials were discontinued. Reverse translational studies in mice demonstrated that IACS-010759 induced behavioral and physiological changes indicative of peripheral neuropathy, which were minimized with the coadministration of a histone deacetylase 6 inhibitor. Additional studies are needed to elucidate the association between OXPHOS inhibition and neurotoxicity, and caution is warranted in the continued development of complex I inhibitors as antitumor agents.
Collapse
Affiliation(s)
- Timothy A Yap
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Naval Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mikhila Mahendra
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jixiang Zhang
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meghan E Collins
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Maria Emilia Di Francesco
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ecaterina E Dumbrava
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sisi Gao
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason P Gay
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonal Gera
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Han
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias J Jabbour
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Jennifer R Molina
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalia Baran
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maro Ohanian
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carolina Salguero
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anand K Singh
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Quanyun A Xu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Zhang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Li
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christopher A Bristow
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meenakshi B Bhattacharjee
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA
| | - Timothy P Heffernan
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher P Vellano
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip Jones
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cobi J Heijnen
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Psychological Sciences, Rice University, Houston, TX, USA
| | - Annemieke Kavelaars
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph R Marszalek
- Translational Research to AdvanCe Therapeutics and Innovation in ONcology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Marina Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
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: 13] [Impact Index Per Article: 4.3] [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
|
8
|
α-Synuclein molecular behavior and nigral proteomic profiling distinguish subtypes of Lewy body disorders. Acta Neuropathol 2022; 144:167-185. [PMID: 35748929 DOI: 10.1007/s00401-022-02453-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/01/2022]
Abstract
Lewy body disorders (LBD), characterized by the deposition of misfolded α-synuclein (α-Syn), are clinically heterogeneous. Although the distribution of α-Syn correlates with the predominant clinical features, the burden of pathology does not fully explain the observed variability in clinical presentation and rate of disease progression. We hypothesized that this heterogeneity might reflect α-Syn molecular diversity, between both patients and different brain regions. Using an ultra-sensitive assay, we evaluated α-Syn seeding in 8 brain regions from 30 LBD patients with different clinical phenotypes and disease durations. Comparing seeding across the clinical phenotypes revealed that hippocampal α-Syn from patients with a cognitive-predominant phenotype had significantly higher seeding capacity than that derived from patients with a motor-predominant phenotype, whose nigral-derived α-Syn in turn had higher seeding capacity than that from cognitive-predominant patients. Interestingly, α-Syn from patients with rapid disease progression (< 3 years to development of advanced disease) had the highest nigral seeding capacity of all the patients included. To validate these findings and explore factors underlying seeding heterogeneity, we performed in vitro toxicity assays, and detailed neuropathological and biochemical examinations. Furthermore, and for the first time, we performed a proteomic-wide profiling of the substantia nigra from 5 high seeder and 5 low seeder patients. The proteomic data suggests a significant disruption in mitochondrial function and lipid metabolism in high seeder cases compared to the low seeders. These observations suggest that distinct molecular populations of α-Syn may contribute to heterogeneity in phenotypes and progression rates in LBD and imply that effective therapeutic strategies might need to be directed at an ensemble of differently misfolded α-Syn species, with the relative contribution of their differing impacts accounting for heterogeneity in the neurodegenerative process.
Collapse
|
9
|
O'Hanlon ME, Tweedy C, Scialo F, Bass R, Sanz A, Smulders-Srinivasan TK. Mitochondrial electron transport chain defects modify Parkinson's disease phenotypes in a Drosophila model. Neurobiol Dis 2022; 171:105803. [PMID: 35764292 DOI: 10.1016/j.nbd.2022.105803] [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: 03/10/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Mitochondrial defects have been implicated in Parkinson's disease (PD) since complex I poisons were found to cause accelerated parkinsonism in young people in the early 1980s. More evidence of mitochondrial involvement arose when many of the genes whose mutations caused inherited PD were discovered to be subcellularly localized to mitochondria or have mitochondrial functions. However, the details of how mitochondrial dysfunction might impact or cause PD remain unclear. The aim of our study was to better understand mitochondrial dysfunction in PD by evaluating mitochondrial respiratory complex mutations in a Drosophila melanogaster (fruit fly) model of PD. METHODS We have conducted a targeted heterozygous enhancer/suppressor screen using Drosophila mutations within mitochondrial electron transport chain (ETC) genes against a null PD mutation in parkin. The interactions were assessed by climbing assays at 2-5 days as an indicator of motor function. A strong enhancer mutation in COX5A was examined further for L-dopa rescue, oxygen consumption, mitochondrial content, and reactive oxygen species. A later timepoint of 16-20 days was also investigated for both COX5A and a suppressor mutation in cyclope. Generalized Linear Models and similar statistical tests were used to verify significance of the findings. RESULTS We have discovered that mutations in individual genes for subunits within the mitochondrial respiratory complexes have interactions with parkin, while others do not, irrespective of complex. One intriguing mutation in a complex IV subunit (cyclope) shows a suppressor rescue effect at early time points, improving the gross motor defects caused by the PD mutation, providing a strong candidate for drug discovery. Most mutations, however, show varying degrees of enhancement or slight suppression of the PD phenotypes. Thus, individual mitochondrial mutations within different oxidative phosphorylation complexes have different interactions with PD with regard to degree and direction. Upon further investigation of the strongest enhancer (COX5A), the mechanism by which these interactions occur initially does not appear to be based on defects in ATP production, but rather may be related to increased levels of reactive oxygen species. CONCLUSIONS Our work highlights some key subunits potentially involved in mechanisms underlying PD pathogenesis, implicating ETC complexes other than complex I in PD.
Collapse
Affiliation(s)
- Maria E O'Hanlon
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, Darlington DL1 1HG, United Kingdom. M.O'
| | - Clare Tweedy
- Biosciences Institute, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK.
| | - Filippo Scialo
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, United Kingdom.
| | - Rosemary Bass
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK.
| | - Alberto Sanz
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, United Kingdom.
| | - Tora K Smulders-Srinivasan
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, Darlington DL1 1HG, United Kingdom; Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK.
| |
Collapse
|
10
|
Therapeutic Targets for Regulating Oxidative Damage Induced by Ischemia-Reperfusion Injury: A Study from a Pharmacological Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8624318. [PMID: 35450409 PMCID: PMC9017553 DOI: 10.1155/2022/8624318] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 12/22/2022]
Abstract
Ischemia-reperfusion (I-R) injury is damage caused by restoring blood flow into ischemic tissues or organs. This complex and characteristic lesion accelerates cell death induced by signaling pathways such as apoptosis, necrosis, and even ferroptosis. In addition to the direct association between I-R and the release of reactive oxygen species and reactive nitrogen species, it is involved in developing mitochondrial oxidative damage. Thus, its mechanism plays a critical role via reactive species scavenging, calcium overload modulation, electron transport chain blocking, mitochondrial permeability transition pore activation, or noncoding RNA transcription. Other receptors and molecules reduce tissue and organ damage caused by this pathology and other related diseases. These molecular targets have been gradually discovered and have essential roles in I-R resolution. Therefore, the current study is aimed at highlighting the importance of these discoveries. In this review, we inquire about the oxidative damage receptors that are relevant to reducing the damage induced by oxidative stress associated with I-R. Several complications on surgical techniques and pathology interventions do not mitigate the damage caused by I-R. Nevertheless, these therapies developed using alternative targets could work as coadjuvants in tissue transplants or I-R-related pathologies
Collapse
|
11
|
Finnegan LK, Chadderton N, Kenna PF, Palfi A, Carty M, Bowie AG, Millington-Ward S, Farrar GJ. SARM1 Ablation Is Protective and Preserves Spatial Vision in an In Vivo Mouse Model of Retinal Ganglion Cell Degeneration. Int J Mol Sci 2022; 23:ijms23031606. [PMID: 35163535 PMCID: PMC8835928 DOI: 10.3390/ijms23031606] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
The challenge of developing gene therapies for genetic forms of blindness is heightened by the heterogeneity of these conditions. However, mechanistic commonalities indicate key pathways that may be targeted in a gene-independent approach. Mitochondrial dysfunction and axon degeneration are common features of many neurodegenerative conditions including retinal degenerations. Here we explore the neuroprotective effect afforded by the absence of sterile alpha and Toll/interleukin-1 receptor motif-containing 1 (SARM1), a prodegenerative NADase, in a rotenone-induced mouse model of retinal ganglion cell loss and visual dysfunction. Sarm1 knockout mice retain visual function after rotenone insult, displaying preservation of photopic negative response following rotenone treatment in addition to significantly higher optokinetic response measurements than wild type mice following rotenone. Protection of spatial vision is sustained over time in both sexes and is accompanied by increased RGC survival and additionally preservation of axonal density in optic nerves of Sarm1−/− mice insulted with rotenone. Primary fibroblasts extracted from Sarm1−/− mice demonstrate an increased oxygen consumption rate relative to those from wild type mice, with significantly higher basal, maximal and spare respiratory capacity. Collectively, our data indicate that Sarm1 ablation increases mitochondrial bioenergetics and confers histological and functional protection in vivo in the mouse retina against mitochondrial dysfunction, a hallmark of many neurodegenerative conditions including a variety of ocular disorders.
Collapse
Affiliation(s)
- Laura K. Finnegan
- Department of Genetics, The School of Genetics and Microbiology, Trinity College Dublin, D02 VF25 Dublin, Ireland; (N.C.); (P.F.K.); (A.P.); (S.M.-W.); (G.J.F.)
- Correspondence:
| | - Naomi Chadderton
- Department of Genetics, The School of Genetics and Microbiology, Trinity College Dublin, D02 VF25 Dublin, Ireland; (N.C.); (P.F.K.); (A.P.); (S.M.-W.); (G.J.F.)
| | - Paul F. Kenna
- Department of Genetics, The School of Genetics and Microbiology, Trinity College Dublin, D02 VF25 Dublin, Ireland; (N.C.); (P.F.K.); (A.P.); (S.M.-W.); (G.J.F.)
- The Research Foundation, Royal Victoria Eye and Ear Hospital, D02 XK51 Dublin, Ireland
| | - Arpad Palfi
- Department of Genetics, The School of Genetics and Microbiology, Trinity College Dublin, D02 VF25 Dublin, Ireland; (N.C.); (P.F.K.); (A.P.); (S.M.-W.); (G.J.F.)
| | - Michael Carty
- Trinity Biomedical Sciences Institute, The School of Biochemistry and Immunology, Trinity College Dublin, D02 R590 Dublin, Ireland; (M.C.); (A.G.B.)
| | - Andrew G. Bowie
- Trinity Biomedical Sciences Institute, The School of Biochemistry and Immunology, Trinity College Dublin, D02 R590 Dublin, Ireland; (M.C.); (A.G.B.)
| | - Sophia Millington-Ward
- Department of Genetics, The School of Genetics and Microbiology, Trinity College Dublin, D02 VF25 Dublin, Ireland; (N.C.); (P.F.K.); (A.P.); (S.M.-W.); (G.J.F.)
| | - G. Jane Farrar
- Department of Genetics, The School of Genetics and Microbiology, Trinity College Dublin, D02 VF25 Dublin, Ireland; (N.C.); (P.F.K.); (A.P.); (S.M.-W.); (G.J.F.)
| |
Collapse
|
12
|
Nm23-H1 activator phenylbutenoid dimer exerts cytotoxic effects on metastatic breast cancer cells by inducing mitochondrial dysfunction only under glucose starvation. Sci Rep 2021; 11:23549. [PMID: 34876614 PMCID: PMC8651694 DOI: 10.1038/s41598-021-02729-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) has become an attractive target in anti-cancer studies in recent years. In this study, we found that a small molecule phenylbutenoid dimer NMac1 (Nm23-H1 activator 1), (±)-trans-3-(3,4-dimethoxyphenyl)-4-[(E)-3,4-dimethoxystyryl]cyclohex-1-ene, a previously identified anti-metastatic agent, has novel anti-proliferative effect only under glucose starvation in metastatic breast cancer cells. NMac1 causes significant activation of AMPK by decreasing ATP synthesis, lowers mitochondrial membrane potential (MMP, ΔΨm), and inhibits oxygen consumption rate (OCR) under glucose starvation. These effects of NMac1 are provoked by a consequence of OXPHOS complex I inhibition. Through the structure–activity relationship (SAR) study of NMac1 derivatives, NMac24 was identified as the most effective compound in anti-proliferation. NMac1 and NMac24 effectively suppress cancer cell proliferation in 3D-spheroid in vivo-like models only under glucose starvation. These results suggest that NMac1 and NMac24 have the potential as anti-cancer agents having cytotoxic effects selectively in glucose restricted cells.
Collapse
|
13
|
Chen J, Li M, Zhou X, Xie A, Cai Z, Fu C, Peng Y, Zhang H, Liu L. Rotenone-Induced Neurodegeneration Is Enabled by a p38-Parkin-ROS Signaling Feedback Loop. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13942-13952. [PMID: 34779196 DOI: 10.1021/acs.jafc.1c04190] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Rotenone, a component of pesticides, is widely used in agriculture and potentially causes Parkinson's disease (PD). However, the regulatory mechanisms of rotenone-induced PD are unclear. Here, we revealed a novel feedback mechanism of p38-Parkin-ROS regulating rotenone-induced PD. Rotenone treatment led to not only the activation of p38 but also Parkin inactivation and reactive oxygen species (ROS) overproduction in SN4741 cells. Meanwhile, p38 activation regulated Parkin phosphorylation at serine 131 to disrupt Parkin-mediated mitophagy. Notably, both p38 inhibition and Parkin overexpression decreased ROS levels. Additionally, the ROS inhibitor N-acetyl-l-cysteine (NAC) inhibited p38 and activated Parkin-mediated mitophagy. Both p38 inhibition and the ROS inhibitor NAC exerted a protective effect by restoring cell death and mitochondrial function in rotenone-induced PD models. Based on these results, the p38-Parkin-ROS signaling pathway is involved in neurodegeneration. This pathway represents a valuable treatment strategy for rotenone-induced PD, and our study provides basic research evidence for the safe use of rotenone in agriculture.
Collapse
Affiliation(s)
- Jialong Chen
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| | - Mingque Li
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| | - Xueqiong Zhou
- Department of Occupational Health and Occupational Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, Guangdong, P. R. China
| | - Ailun Xie
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| | - Ziwei Cai
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| | - Chunlai Fu
- Department of Emergency Intensive Care Unit, Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan 523808, Guangdong, P. R. China
| | - Yongming Peng
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| | - He Zhang
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| | - Linhua Liu
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, P. R. China
| |
Collapse
|
14
|
Amoedo ND, Sarlak S, Obre E, Esteves P, Bégueret H, Kieffer Y, Rousseau B, Dupis A, Izotte J, Bellance N, Dard L, Redonnet-Vernhet I, Punzi G, Rodrigues MF, Dumon E, Mafhouf W, Guyonnet-Dupérat V, Gales L, Palama T, Bellvert F, Dugot-Senan N, Claverol S, Baste JM, Lacombe D, Rezvani HR, Pierri CL, Mechta-Grigoriou F, Thumerel M, Rossignol R. Targeting the mitochondrial trifunctional protein restrains tumor growth in oxidative lung carcinomas. J Clin Invest 2021; 131:133081. [PMID: 33393495 DOI: 10.1172/jci133081] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
Metabolic reprogramming is a common hallmark of cancer, but a large variability in tumor bioenergetics exists between patients. Using high-resolution respirometry on fresh biopsies of human lung adenocarcinoma, we identified 2 subgroups reflected in the histologically normal, paired, cancer-adjacent tissue: high (OX+) mitochondrial respiration and low (OX-) mitochondrial respiration. The OX+ tumors poorly incorporated [18F]fluorodeoxy-glucose and showed increased expression of the mitochondrial trifunctional fatty acid oxidation enzyme (MTP; HADHA) compared with the paired adjacent tissue. Genetic inhibition of MTP altered OX+ tumor growth in vivo. Trimetazidine, an approved drug inhibitor of MTP used in cardiology, also reduced tumor growth and induced disruption of the physical interaction between the MTP and respiratory chain complex I, leading to a cellular redox and energy crisis. MTP expression in tumors was assessed using histology scoring methods and varied in negative correlation with [18F]fluorodeoxy-glucose incorporation. These findings provide proof-of-concept data for preclinical, precision, bioenergetic medicine in oxidative lung carcinomas.
Collapse
Affiliation(s)
- Nivea Dias Amoedo
- CELLOMET, Bordeaux, France.,INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Saharnaz Sarlak
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Emilie Obre
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Pauline Esteves
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Hugues Bégueret
- Bordeaux University, Bordeaux, France.,Pathology Department, Haut-Lévèque Hospital, CHU Bordeaux, Bordeaux, France
| | - Yann Kieffer
- Stress and Cancer Laboratory, Curie Institute - INSERM U830, Paris, France
| | - Benoît Rousseau
- INSERM U1211, Bordeaux, France.,Transgenic Animal Facility A2, University of Bordeaux, Bordeaux, France
| | - Alexis Dupis
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Julien Izotte
- INSERM U1211, Bordeaux, France.,Transgenic Animal Facility A2, University of Bordeaux, Bordeaux, France
| | - Nadège Bellance
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Laetitia Dard
- CELLOMET, Bordeaux, France.,INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Isabelle Redonnet-Vernhet
- CELLOMET, Bordeaux, France.,INSERM U1211, Bordeaux, France.,Biochemistry Department, Pellegrin Hospital, CHU Bordeaux, Bordeaux, France
| | - Giuseppe Punzi
- Laboratory of Biochemistry and Molecular Biology, University of Bari,Bari, Italy
| | | | - Elodie Dumon
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | - Walid Mafhouf
- Bordeaux University, Bordeaux, France.,INSERM U1035, Bordeaux, France
| | | | - Lara Gales
- Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés (LISBP), Institut National des Sciences Appliquées (INSA)/CNRS 5504 - UMR INSA/Institut National de la Recherche Agronomique (INRA) 792, Toulouse, France
| | - Tony Palama
- Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés (LISBP), Institut National des Sciences Appliquées (INSA)/CNRS 5504 - UMR INSA/Institut National de la Recherche Agronomique (INRA) 792, Toulouse, France
| | - Floriant Bellvert
- Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés (LISBP), Institut National des Sciences Appliquées (INSA)/CNRS 5504 - UMR INSA/Institut National de la Recherche Agronomique (INRA) 792, Toulouse, France
| | | | - Stéphane Claverol
- Bordeaux University, Bordeaux, France.,Functional Genomics Center (CGFB), Proteomics Facility, Bordeaux, France
| | - Jean-Marc Baste
- Thoracic Surgery, Haut-Lévèque Hospital, CHU Bordeaux, Bordeaux, France
| | - Didier Lacombe
- INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| | | | - Ciro Leonardo Pierri
- Laboratory of Biochemistry and Molecular Biology, University of Bari,Bari, Italy
| | | | - Matthieu Thumerel
- Thoracic Surgery, Haut-Lévèque Hospital, CHU Bordeaux, Bordeaux, France
| | - Rodrigue Rossignol
- CELLOMET, Bordeaux, France.,INSERM U1211, Bordeaux, France.,Bordeaux University, Bordeaux, France
| |
Collapse
|
15
|
Leggio L, Paternò G, Vivarelli S, Falzone GG, Giachino C, Marchetti B, Iraci N. Extracellular Vesicles as Novel Diagnostic and Prognostic Biomarkers for Parkinson's Disease. Aging Dis 2021; 12:1494-1515. [PMID: 34527424 PMCID: PMC8407885 DOI: 10.14336/ad.2021.0527] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 05/27/2021] [Indexed: 12/29/2022] Open
Abstract
The elderly population will significantly increase in the next decade and, with it, the proportion of people affected by age-related diseases. Among them, one of the most invalidating is Parkinson's disease (PD), characterized by motor- and non-motor dysfunctions which strongly impair the quality of life of affected individuals. PD is characterized by the progressive degeneration of dopaminergic neurons, with consequent dopamine depletion, and the accumulation of misfolded α-synuclein aggregates. Although 150 years have passed since PD first description, no effective therapies are currently available, but only palliative treatments. Importantly, PD is often diagnosed when the neuronal loss is elevated, making difficult any therapeutic intervention. In this context, two key challenges remain unanswered: (i) the early diagnosis to avoid the insurgence of irreversible symptoms; and (ii) the reliable monitoring of therapy efficacy. Research strives to identify novel biomarkers for PD diagnosis, prognosis, and therapeutic follow-up. One of the most promising sources of biomarkers is represented by extracellular vesicles (EVs), a heterogeneous population of nanoparticles, released by all cells in the microenvironment. Brain-derived EVs are able to cross the blood-brain barrier, protecting their payload from enzymatic degradation, and are easily recovered from biofluids. Interestingly, EV content is strongly influenced by the specific pathophysiological status of the donor cell. In this manuscript, the role of EVs as source of novel PD biomarkers is discussed, providing all recent findings concerning relevant proteins and miRNAs carried by PD patient-derived EVs, from several biological specimens. Moreover, the contribution of mitochondria-derived EVs will be dissected. Finally, the promising possibility to use EVs as source of markers to monitor PD therapy efficacy will be also examined. In the future, larger cohort studies will help to validate these EV-associated candidates, that might be effectively used as non-invasive and robust source of biomarkers for PD.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Giovanna G Falzone
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| | - Carmela Giachino
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy.
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy.
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, 95125 Catania, Italy.
| |
Collapse
|
16
|
Duarte JN. Neuroinflammatory Mechanisms of Mitochondrial Dysfunction and Neurodegeneration in Glaucoma. J Ophthalmol 2021; 2021:4581909. [PMID: 33953963 PMCID: PMC8064803 DOI: 10.1155/2021/4581909] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/29/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
The exact mechanism of retinal ganglion cell loss in the pathogenesis of glaucoma is yet to be understood. Mitochondrial damage-associated molecular patterns (DAMPs) resulting from mitochondrial dysfunction have been linked to Leber's hereditary optic neuropathy and autosomal dominant optic atrophy, as well as to brain neurodegenerative diseases. Recent evidence shows that, in conditions where mitochondria are damaged, a sustained inflammatory response and downstream pathological inflammation may ensue. Mitochondrial damage has been linked to the accumulation of age-related mitochondrial DNA mutations and mitochondrial dysfunction, possibly through aberrant reactive oxygen species production and defective mitophagy. The present review focuses on how mitochondrial dysfunction may overwhelm the ability of neurons and glial cells to adequately maintain homeostasis and how mitochondria-derived DAMPs trigger the immune system and induce neurodegeneration.
Collapse
Affiliation(s)
- Joao N. Duarte
- Neuroinflammation Unit, Biotech Research & Innovation Center, University of Copenhagen, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Section 7631, Rigshospitalet, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Bertrand L, Velichkovska M, Toborek M. Cerebral Vascular Toxicity of Antiretroviral Therapy. J Neuroimmune Pharmacol 2021; 16:74-89. [PMID: 31209776 PMCID: PMC7952282 DOI: 10.1007/s11481-019-09858-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/27/2019] [Indexed: 01/14/2023]
Abstract
HIV infection is associated with comorbidities that are likely to be driven not only by HIV itself, but also by the toxicity of long-term use of antiretroviral therapy (ART). Indeed, increasing evidence demonstrates that the antiretroviral drugs used for HIV treatment have toxic effects resulting in various cellular and tissue pathologies. The blood-brain barrier (BBB) is a modulated anatomophysiological interface which separates and controls substance exchange between the blood and the brain parenchyma; therefore, it is particularly exposed to ART-induced toxicity. Balancing the health risks and gains of ART has to be considered in order to maximize the positive effects of therapy. The current review discusses the cerebrovascular toxicity of ART, with the focus on mitochondrial dysfunction. Graphical Abstract Graphical representation of the interactions between HIV, antiretroviral therapy (ART), and the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Luc Bertrand
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA
| | - Martina Velichkovska
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA.
| |
Collapse
|
18
|
Panchal K, Tiwari AK. Mitochondrial dynamics, a key executioner in neurodegenerative diseases. Mitochondrion 2019; 47:151-173. [PMID: 30408594 DOI: 10.1016/j.mito.2018.11.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/08/2018] [Accepted: 11/02/2018] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases (NDs) are the group of disorder that includes brain, peripheral nerves, spinal cord and results in sensory and motor neuron dysfunction. Several studies have shown that mitochondrial dynamics and their axonal transport play a central role in most common NDs such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and Amyotrophic Lateral Sclerosis (ALS) etc. In normal physiological condition, there is a balance between mitochondrial fission and fusion process while any alteration to these processes cause defect in ATP (Adenosine Triphosphate) biogenesis that lead to the onset of several NDs. Also, mitochondria mediated ROS may induce lipid and protein peroxidation, energy deficiency environment in the neurons and results in cell death and defective neurotransmission. Though, mitochondria is a well-studied cell organelle regulating the cellular energy demands but still, its detail role or association in NDs is under observation. In this review, we have summarized an updated mitochondria and their possible role in different NDs with the therapeutic strategy to improve the mitochondrial functions.
Collapse
Affiliation(s)
- Komal Panchal
- Genetics & Developmental Biology Laboratory, School of Biological Sciences & Biotechnology, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar 382426, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, School of Biological Sciences & Biotechnology, Institute of Advanced Research (IAR), Koba, Institutional Area, Gandhinagar 382426, India.
| |
Collapse
|
19
|
Laurencé C, Zeghbib N, Rivard M, Lehri-Boufala S, Lachaise I, Barau C, Le Corvoisier P, Martens T, Garrigue-Antar L, Morin C. A new human pyridinium metabolite of furosemide, inhibitor of mitochondrial complex I, is a candidate inducer of neurodegeneration. Biochem Pharmacol 2019; 160:14-23. [DOI: 10.1016/j.bcp.2018.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/07/2018] [Indexed: 11/29/2022]
|
20
|
Activation of microglia synergistically enhances neurodegeneration caused by MPP + in human SH-SY5Y cells. Eur J Pharmacol 2019; 850:64-74. [PMID: 30684467 DOI: 10.1016/j.ejphar.2019.01.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 01/21/2023]
Abstract
While MPP+ may not directly activate microglia, the initial neuronal damage inflicted by the toxin may trigger microglia, possibly leading to synergistic pro-apoptotic interaction between neuro-inflammation and toxin-induced neurotoxicity, which may further aggravate neurodegeneration. However, what molecular targets are synergistically up or downregulated during this interaction is not well understood. Here, we addressed this by co-culturing fully differentiated human SH-SY5Y cells treated with parkinsonian toxin 1-Methyl-4-phenylpyridinium (MPP+), with endotoxin-activated microglial cell line EOC 20 to determine how this interaction affects pro-apoptotic (p38, JNK, and bax:bcl2 ratios) and pro-survival (NF-κB, MEK1) signaling at both mRNA and protein levels. Concurrent MPP+ and endotoxin-treatment aggravated a decrease in SH-SY5Y cell viability and caused strong synergistic increases in the bax:bcl2 ratio, but also NF-κB and JNK signaling. These effects were attenuated by microglia inhibitor minocycline. Altogether, these data provide further molecular insights into the important role or even conditional requirement of microglia activation in the progressive neurodegenerative nature of PD.
Collapse
|
21
|
Yuan SH, Hiramatsu N, Liu Q, Sun XV, Lenh D, Chan P, Chiang K, Koo EH, Kao AW, Litvan I, Lin JH. Tauopathy-associated PERK alleles are functional hypomorphs that increase neuronal vulnerability to ER stress. Hum Mol Genet 2018; 27:3951-3963. [PMID: 30137327 PMCID: PMC6216228 DOI: 10.1093/hmg/ddy297] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/04/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by tau protein pathology in the nervous system. EIF2AK3 (eukaryotic translation initiation factor 2 alpha kinase 3), also known as PERK (protein kinase R-like endoplasmic reticulum kinase), was identified by genome-wide association study as a genetic risk factor in several tauopathies. PERK is a key regulator of the Unfolded Protein Response (UPR), an intracellular signal transduction mechanism that protects cells from endoplasmic reticulum (ER) stress. PERK variants had previously been identified in Wolcott-Rallison Syndrome, a rare autosomal recessive metabolic disorder, and these variants completely abrogated the function of PERK's kinase domain or prevented PERK expression. In contrast, the PERK tauopathy risk variants were distinct from the Wolcott-Rallison variants and introduced missense alterations throughout the PERK protein. The function of PERK tauopathy variants and their effects on neurodegeneration are unknown. Here, we discovered that tauopathy-associated PERK alleles showed reduced signaling activity and increased PERK protein turnover compared to protective PERK alleles. We found that iPSC-derived neurons carrying PERK risk alleles were highly vulnerable to ER stress-induced injury with increased tau pathology. We found that chemical inhibition of PERK in human iPSC-derived neurons also increased neuronal cell death in response to ER stress. Our results indicate that tauopathy-associated PERK alleles are functional hypomorphs during the UPR. We propose that reduced PERK function leads to neurodegeneration by increasing neuronal vulnerability to ER stress-associated damage. In this view, therapies to enhance PERK signaling would benefit at-risk carriers of hypomorphic alleles.
Collapse
Affiliation(s)
- Shauna H Yuan
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Nobuhiko Hiramatsu
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA
| | - Qing Liu
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Xuehan Victoria Sun
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA
| | - David Lenh
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Priscilla Chan
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA
| | - Karen Chiang
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA
| | - Edward H Koo
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- Departments of Medicine and Physiology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Aimee W Kao
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Irene Litvan
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Jonathan H Lin
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
22
|
Supandi F, van Beek JHGM. Computational prediction of changes in brain metabolic fluxes during Parkinson's disease from mRNA expression. PLoS One 2018; 13:e0203687. [PMID: 30208076 PMCID: PMC6135490 DOI: 10.1371/journal.pone.0203687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 08/24/2018] [Indexed: 12/12/2022] Open
Abstract
Background Parkinson’s disease is a widespread neurodegenerative disorder which affects brain metabolism. Although changes in gene expression during disease are often measured, it is difficult to predict metabolic fluxes from gene expression data. Here we explore the hypothesis that changes in gene expression for enzymes tend to parallel flux changes in biochemical reaction pathways in the brain metabolic network. This hypothesis is the basis of a computational method to predict metabolic flux changes from post-mortem gene expression measurements in Parkinson’s disease (PD) brain. Results We use a network model of central metabolism and optimize the correspondence between relative changes in fluxes and in gene expression. To this end we apply the Least-squares with Equalities and Inequalities algorithm integrated with Flux Balance Analysis (Lsei-FBA). We predict for PD (1) decreases in glycolytic rate and oxygen consumption and an increase in lactate production in brain cortex that correspond with measurements (2) relative flux decreases in ATP synthesis, in the malate-aspartate shuttle and midway in the TCA cycle that are substantially larger than relative changes in glucose uptake in the substantia nigra, dopaminergic neurons and most other brain regions (3) shifts in redox shuttles between cytosol and mitochondria (4) in contrast to Alzheimer’s disease: little activation of the gamma-aminobutyric acid shunt pathway in compensation for decreased alpha-ketoglutarate dehydrogenase activity (5) in the globus pallidus internus, metabolic fluxes are increased, reflecting increased functional activity. Conclusion Our method predicts metabolic changes from gene expression data that correspond in direction and order of magnitude with presently available experimental observations during Parkinson’s disease, indicating that the hypothesis may be useful for some biochemical pathways. Lsei-FBA generates predictions of flux distributions in neurons and small brain regions for which accurate metabolic flux measurements are not yet possible.
Collapse
Affiliation(s)
- Farahaniza Supandi
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| | - Johannes H. G. M. van Beek
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Academic Medical Center, AZ Amsterdam, the Netherlands
| |
Collapse
|
23
|
De Miranda BR, Rocha EM, Bai Q, El Ayadi A, Hinkle D, Burton EA, Timothy Greenamyre J. Astrocyte-specific DJ-1 overexpression protects against rotenone-induced neurotoxicity in a rat model of Parkinson's disease. Neurobiol Dis 2018; 115:101-114. [PMID: 29649621 PMCID: PMC5943150 DOI: 10.1016/j.nbd.2018.04.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/29/2018] [Accepted: 04/06/2018] [Indexed: 02/07/2023] Open
Abstract
DJ-1 is a redox-sensitive protein with several putative functions important in mitochondrial physiology, protein transcription, proteasome regulation, and chaperone activity. High levels of DJ-1 immunoreactivity are reported in astrocytes surrounding pathology associated with idiopathic Parkinson's disease, possibly reflecting the glial response to oxidative damage. Previous studies showed that astrocytic over-expression of DJ-1 in vitro prevented oxidative stress and mitochondrial dysfunction in primary neurons. Based on these observations, we developed a pseudotyped lentiviral gene transfer vector with specific tropism for CNS astrocytes in vivo to overexpress human DJ-1 protein in astroglial cells. Following vector delivery to the substantia nigra and striatum of adult Lewis rats, the DJ-1 transgene was expressed robustly and specifically within astrocytes. There was no observable transgene expression in neurons or other glial cell types. Three weeks after vector infusion, animals were exposed to rotenone to induce Parkinson's disease-like pathology, including loss of dopaminergic neurons, accumulation of endogenous α-synuclein, and neuroinflammation. Animals over-expressing hDJ-1 in astrocytes were protected from rotenone-induced neurodegeneration, and displayed a marked reduction in neuronal oxidative stress and microglial activation. In addition, α-synuclein accumulation and phosphorylation were decreased within substantia nigra dopaminergic neurons in DJ-1-transduced animals, and expression of LAMP-2A, a marker of chaperone mediated autophagy, was increased. Together, these data indicate that astrocyte-specific overexpression of hDJ-1 protects neighboring neurons against multiple pathologic features of Parkinson's disease and provides the first direct evidence in vivo of a cell non-autonomous neuroprotective function of astroglial DJ-1.
Collapse
Affiliation(s)
- Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qing Bai
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Amina El Ayadi
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - David Hinkle
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States.
| |
Collapse
|
24
|
Pinto LC, De Souza CO, De Souza SA, Da Silva HB, Da Silva RR, Cerqueira-Lima AT, Teixeira TO, Da Silva TM, Medeiros KC, Bittencourt M, Brandão HR, Druzian JI, Conceição AS, Lopes MV, Figueiredo CA. Potential of Annona muricata L. seed oil: phytochemical and nutritional characterization associated with non-toxicity. GRASAS Y ACEITES 2018. [DOI: 10.3989/gya.0777171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The aim of this study was to evaluate the nutritional quality, phenolic compounds, fatty acid and antioxidant activity in vitro as well as a toxicological screening of A. muricata seed oil in vivo. The chemical composition and quantification of phenolic compounds were determined by the Adolfo Lutz Institute normative. The antioxidant activity was evaluated by DPPH, FRAP and ABTS methods. The oil was extracted by chloroform/ methanol and precipitated crude (AmPtO) and supernatant oils (AmSO) were obtained. The fatty acid profile was evaluated by gas chromatography and total compounds by HPLC-DAD. BALB/C mice received AmPtO and AmSO (0.5 and 1.0mL·Kg-1) for 14 days. Toxicity parameters were assessed. The major fatty acids in the oil were oleic (39.2%) and linoleic (33%). HPLC-DAD suggested the presence of acetogenins (annonacin: 595 [M-H]-), with a greater presence in AmPtO. The AmPtO group showed toxicity, which may be related to the acetogenin content in AmPtO. The AmSO group showed no toxicity and this oil has potential for food or medicinal use.
Collapse
|
25
|
Clinical effects of chemical exposures on mitochondrial function. Toxicology 2017; 391:90-99. [PMID: 28757096 DOI: 10.1016/j.tox.2017.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/07/2017] [Accepted: 07/17/2017] [Indexed: 12/16/2022]
Abstract
Mitochondria are critical for the provision of ATP for cellular energy requirements. Tissue and organ functions are dependent on adequate ATP production, especially when energy demand is high. Mitochondria also play a role in a vast array of important biochemical pathways including apoptosis, generation and detoxification of reactive oxygen species, intracellular calcium regulation, steroid hormone and heme synthesis, and lipid metabolism. The complexity of mitochondrial structure and function facilitates its diverse roles but also enhances its vulnerability. Primary disorders of mitochondrial bioenergetics, or Primary Mitochondrial Diseases (PMD) are due to inherited genetic defects in the nuclear or mitochondrial genomes that result in defective oxidative phosphorylation capacity and cellular energy production. Secondary mitochondrial dysfunction is observed in a wide range of diseases such as Alzheimer's and Parkinson's disease. Several lines of evidence suggest that environmental exposures cause substantial mitochondrial dysfunction. Whereby literature from experimental and human studies on exposures associated with Alzheimer's and Parkinson's diseases exist, the significance of exposures as potential triggers in Primary Mitochondrial Disease (PMD) is an emerging clinical question that has not been systematically studied.
Collapse
|
26
|
Bonneau N, Cynober T, Jullian JC, Champy P. 1 H qNMR Quantification of Annonaceous Acetogenins in Crude Extracts of Annona muricata L. Fruit Pulp. PHYTOCHEMICAL ANALYSIS : PCA 2017; 28:251-256. [PMID: 28092423 DOI: 10.1002/pca.2668] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 11/01/2016] [Accepted: 11/10/2016] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Annonaceous acetogenins (AAGs) constitute a group of environmental neurotoxins, possibly implicated in sporadic atypical Parkinsonism/dementia complexes. The recent evidencing of complex mixtures of AAGs in edible fruits and derived food products requires efficient and practical analytical tools for an estimation of human exposure. OBJECTIVE To develop a simple method for the direct quantitation of the majority of AAGs (sub-types 1a and 1b) within crude extracts, using commonly available 1 H-NMR spectrometers, for food control. METHODOLOGY Method development was carried out on 400 MHz and 300 MHz spectrometers, for routine application on fruits crude extracts of Annona muricata L. The method was validated with annonacin and squamocin as reference compounds. Two internal standards (ISs), fumaric acid and dimethyl fumarate, were successfully used, in deuterated methanol (CD3 OD) and deuterated chloroform (CDCl3 ), respectively. RESULTS Quantitation was carried out using signals corresponding to the deshielded ethylenic protons characterising most AAGs, at δ 7.18 or δ 6.98 ppm in CDCl3 . The limit of quantification (LOQ) was 2.5 mM, with acceptable accuracy, and the limit of detection (LOD) was 0.5 mM. The AAGs contents measured in seven distinct fruit samples of Annona muricata ranged from 14 μmol to 226 μmol of AAGs per 100 g fresh pulp (i.e. 0.14 mmol to 1.3 mmol of AAGs per fruit). CONCLUSION A simple, accurate and specific method for quantification of AAGs content was developed and validated for routine application to fruit pulp crude extracts. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Natacha Bonneau
- Laboratoire de Pharmacognosie, BioCIS, Univ. Paris-Sud, CNRS, Université Paris-Saclay, UFR Pharmacie, Châtenay-Malabry, France
| | - Timothé Cynober
- Laboratoire de Pharmacognosie, BioCIS, Univ. Paris-Sud, CNRS, Université Paris-Saclay, UFR Pharmacie, Châtenay-Malabry, France
| | - Jean-Christophe Jullian
- Laboratoire de Pharmacognosie, BioCIS, Univ. Paris-Sud, CNRS, Université Paris-Saclay, UFR Pharmacie, Châtenay-Malabry, France
| | - Pierre Champy
- Laboratoire de Pharmacognosie, BioCIS, Univ. Paris-Sud, CNRS, Université Paris-Saclay, UFR Pharmacie, Châtenay-Malabry, France
| |
Collapse
|
27
|
Ferramosca A, Di Giacomo M, Zara V. Antioxidant dietary approach in treatment of fatty liver: New insights and updates. World J Gastroenterol 2017; 23:4146-4157. [PMID: 28694655 PMCID: PMC5483489 DOI: 10.3748/wjg.v23.i23.4146] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/22/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common clinicopathological condition, encompassing a range of conditions caused by lipid deposition within liver cells. To date, no approved drugs are available for the treatment of NAFLD, despite the fact that it represents a serious and growing clinical problem in the Western world. Identification of the molecular mechanisms leading to NAFLD-related fat accumulation, mitochondrial dysfunction and oxidative balance impairment facilitates the development of specific interventions aimed at preventing the progression of hepatic steatosis. In this review, we focus our attention on the role of dysfunctions in mitochondrial bioenergetics in the pathogenesis of fatty liver. Major data from the literature about the mitochondrial targeting of some antioxidant molecules as a potential treatment for hepatic steatosis are described and critically analysed. There is ample evidence of the positive effects of several classes of antioxidants, such as polyphenols (i.e., resveratrol, quercetin, coumestrol, anthocyanins, epigallocatechin gallate and curcumin), carotenoids (i.e., lycopene, astaxanthin and fucoxanthin) and glucosinolates (i.e., glucoraphanin, sulforaphane, sinigrin and allyl-isothiocyanate), on the reversion of fatty liver. Although the mechanism of action is not yet fully elucidated, in some cases an indirect interaction with mitochondrial metabolism is expected. We believe that such knowledge will eventually translate into the development of novel therapeutic approaches for fatty liver.
Collapse
|
28
|
Catastrophic cliffs: a partial suggestion for selective vulnerability in neurodegenerative diseases. Biochem Soc Trans 2016; 44:659-61. [PMID: 27068985 DOI: 10.1042/bst20150287] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Indexed: 01/03/2023]
Abstract
For each of the neurodegenerative syndromes, we now know many pathogenic and/or causative genetic risk loci. Here, I suggest that this wealth of knowledge now allows us to start to understand what are the specific vulnerabilities of different neuronal types and to suggest that each neuronal type is closer to different modes of catastrophic failure. I suggest that this is part of the basis for selective vulnerability in neurodegenerative disease.
Collapse
|
29
|
Protein undernutrition during development and oxidative impairment in the central nervous system (CNS): potential factors in the occurrence of metabolic syndrome and CNS disease. J Dev Orig Health Dis 2016; 7:513-524. [DOI: 10.1017/s2040174416000246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondria play a regulatory role in several essential cell processes including cell metabolism, calcium balance and cell viability. In recent years, it has been postulated that mitochondria participate in the pathogenesis of a number of chronic diseases, including central nervous system disorders. Thus, the concept of mitochondrial function now extends far beyond the common view of this organelle as the ‘powerhouse’ of the cell to a new appreciation of the mitochondrion as a transducer of early metabolic insult into chronic disease in later life. In this review, we have attempted to describe some of the associations between nutritional status and mitochondrial function (and dysfunction) during embryonic development with the occurrence of neural oxidative imbalance and neurogenic disease in adulthood.
Collapse
|
30
|
Guerra de Souza AC, Prediger RD, Cimarosti H. SUMO-regulated mitochondrial function in Parkinson's disease. J Neurochem 2016; 137:673-86. [PMID: 26932327 DOI: 10.1111/jnc.13599] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/15/2016] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder characterized by cardinal motor signs such as rigidity, bradykinesia or rest tremor that arise from a significant death of dopaminergic neurons. Non-dopaminergic degeneration also occurs and it seems to induce the deficits in olfactory, emotional, and memory functions that precede the classical motor symptoms in PD. Despite the majority of PD cases being sporadic, several genes have previously been associated with the hereditary forms of the disease. The proteins encoded by some of these genes, including α-synuclein, DJ-1, and parkin, are modified by small ubiquitin-like modifier (SUMO), a post-translational modification that regulates a variety of cellular processes. Among the several pathogenic mechanisms proposed for PD is mitochondrial dysfunction. Recent studies suggest that SUMOylation can interfere with mitochondrial dynamics, which is essential for neuronal function, and may play a pivotal role in PD pathogenesis. Here, we present an overview of recent studies on mitochondrial disturbance in PD and the potential SUMO-modified proteins and pathways involved in this process. SUMOylation, a post-translational modification, interferes with mitochondrial dynamics, and may play a pivotal role in Parkinson's disease (PD). SUMOylation maintains α-synuclein (α-syn) in a soluble form and activates DJ-1, decreasing mitochondrial oxidative stress. SUMOylation may reduce the amount of parkin available for mitochondrial recruitment and decreases mitochondrial biogenesis through suppression of peroxisomal proliferator-activated receptor-γ co-activator 1 α (PGC-1α). Mitochondrial fission can be regulated by dynamin-related protein 1 SUMO-1- or SUMO-2/3-ylation. A fine balance for the SUMOylation/deSUMOylation of these proteins is required to ensure adequate mitochondrial function in PD.
Collapse
Affiliation(s)
- Ana Cristina Guerra de Souza
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Trindade, Florianópolis, SC, Brazil
| | - Rui Daniel Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Trindade, Florianópolis, SC, Brazil
| | - Helena Cimarosti
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Campus Trindade, Florianópolis, SC, Brazil
| |
Collapse
|
31
|
Bonneau N, Schmitz-Afonso I, Brunelle A, Touboul D, Champy P. Quantification of the environmental neurotoxin annonacin in Rat brain by UPLC-MS/MS. Toxicon 2016; 118:129-33. [PMID: 27058514 DOI: 10.1016/j.toxicon.2016.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 10/22/2022]
Abstract
The Annonaceous acetogenin annonacin is an environmental neurotoxin identified in the pulp of several fruits of the Annonaceae family, whose consumption was linked to the occurrence of sporadic atypical Parkinsonism with dementia. A method for its quantification in Rat brain homogenates by UPLC-MS/MS in selected reaction monitoring (SRM) mode was developed and validated. This method was applied to the quantitation of annonacin in Rat brain after intravenous (0.5 mg/kg) and oral (10 mg/kg, 100 mg/kg) administration.
Collapse
Affiliation(s)
- Natacha Bonneau
- Laboratoire de Pharmacognosie, BioCIS, Univ. Paris-Sud, CNRS, Université Paris-Saclay, UFR Pharmacie, 5 rue J.-B. Clément, 92290, Châtenay-Malabry, France
| | - Isabelle Schmitz-Afonso
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Alain Brunelle
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - David Touboul
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Saclay, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Pierre Champy
- Laboratoire de Pharmacognosie, BioCIS, Univ. Paris-Sud, CNRS, Université Paris-Saclay, UFR Pharmacie, 5 rue J.-B. Clément, 92290, Châtenay-Malabry, France.
| |
Collapse
|
32
|
Duda J, Pötschke C, Liss B. Converging roles of ion channels, calcium, metabolic stress, and activity pattern of Substantia nigra dopaminergic neurons in health and Parkinson's disease. J Neurochem 2016; 139 Suppl 1:156-178. [PMID: 26865375 PMCID: PMC5095868 DOI: 10.1111/jnc.13572] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 12/18/2022]
Abstract
Dopamine‐releasing neurons within the Substantia nigra (SN DA) are particularly vulnerable to degeneration compared to other dopaminergic neurons. The age‐dependent, progressive loss of these neurons is a pathological hallmark of Parkinson's disease (PD), as the resulting loss of striatal dopamine causes its major movement‐related symptoms. SN DA neurons release dopamine from their axonal terminals within the dorsal striatum, and also from their cell bodies and dendrites within the midbrain in a calcium‐ and activity‐dependent manner. Their intrinsically generated and metabolically challenging activity is created and modulated by the orchestrated function of different ion channels and dopamine D2‐autoreceptors. Here, we review increasing evidence that the mechanisms that control activity patterns and calcium homeostasis of SN DA neurons are not only crucial for their dopamine release within a physiological range but also modulate their mitochondrial and lysosomal activity, their metabolic stress levels, and their vulnerability to degeneration in PD. Indeed, impaired calcium homeostasis, lysosomal and mitochondrial dysfunction, and metabolic stress in SN DA neurons represent central converging trigger factors for idiopathic and familial PD. We summarize double‐edged roles of ion channels, activity patterns, calcium homeostasis, and related feedback/feed‐forward signaling mechanisms in SN DA neurons for maintaining and modulating their physiological function, but also for contributing to their vulnerability in PD‐paradigms. We focus on the emerging roles of maintained neuronal activity and calcium homeostasis within a physiological bandwidth, and its modulation by PD‐triggers, as well as on bidirectional functions of voltage‐gated L‐type calcium channels and metabolically gated ATP‐sensitive potassium (K‐ATP) channels, and their probable interplay in health and PD.
We propose that SN DA neurons possess several feedback and feed‐forward mechanisms to protect and adapt their activity‐pattern and calcium‐homeostasis within a physiological bandwidth, and that PD‐trigger factors can narrow this bandwidth. We summarize roles of ion channels in this view, and findings documenting that both, reduced as well as elevated activity and associated calcium‐levels can trigger SN DA degeneration.
This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Johanna Duda
- Department of Applied Physiology, Ulm University, Ulm, Germany
| | | | - Birgit Liss
- Department of Applied Physiology, Ulm University, Ulm, Germany.
| |
Collapse
|
33
|
Yu G, Deng A, Tang W, Ma J, Yuan C, Ma J. Hydroxytyrosol induces phase II detoxifying enzyme expression and effectively protects dopaminergic cells against dopamine- and 6-hydroxydopamine induced cytotoxicity. Neurochem Int 2016; 96:113-20. [PMID: 26970393 DOI: 10.1016/j.neuint.2016.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/22/2016] [Accepted: 03/06/2016] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is the second most common late-age onset neurodegenerative disease. Except for the symptomatic alleviating treatment, no disease modifying therapy is currently available. In this study, we investigated the potential neuroprotective role of hydroxytyrosol (HT), a major phenolic compound present in olive oil, against dopaminergic cell death. We found that HT effectively protected dopaminergic SH-SY5Y cells against dopamine (DA) and 6-hydroxydopamine (6-OHDA) induced cell death, but had no apparent effect on 1-methyl-4-phenylpyridinium (MPP(+))-induced cytotoxicity. Furthermore, we have shown that HT efficiently induced the expression of phase II detoxifying enzymes, including NAD(P)H quinone oxidoreductase 1 (NQO1). Using an NQO1 inhibitor, we revealed that increased NQO1 expression contributed to the protective effect of HT against dopaminergic cell death. Together, our findings suggest that HT has a protective effect against DA- and 6-OHDA-induced dopaminergic cell death, supporting the beneficial effect of olive oil in preventing DA-metabolism related dopaminergic neuron dysfunction.
Collapse
Affiliation(s)
- Guohua Yu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, Shanghai, China.
| | - Ajun Deng
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, Shanghai, China
| | - Wanbin Tang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, Shanghai, China
| | - Junzhi Ma
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, Shanghai, China
| | - Chonggang Yuan
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiyan Ma
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, Shanghai, China; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| |
Collapse
|
34
|
Legradi J, el Abdellaoui N, van Pomeren M, Legler J. Comparability of behavioural assays using zebrafish larvae to assess neurotoxicity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:16277-89. [PMID: 25399529 DOI: 10.1007/s11356-014-3805-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 11/02/2014] [Indexed: 05/25/2023]
Abstract
Testing of compounds for neurotoxicity has become increasingly important in recent years. It has been shown that neurological disorders like autism may be related to chemical exposures, which may play a crucial role in the progression of these diseases. Special attention has been be given to the substances causing developmental neurotoxicity as the developing nervous system is more vulnerable to impacts by chemicals than the adult nervous system. The zebrafish (Danio rerio) is a well-established model species in developmental biology and an emerging model in behavioural and neurological studies. Zebrafish larvae display numerous behavioural patterns highly similar to rodents and humans. Their physical characteristics make them well suited for automated high-throughput screening. In the last years, the number of behavioural studies conducted with zebrafish larvae has increased notably. The goal of this review is to provide an overview of behavioural assays commonly used to test substances for developmental neurotoxicity. Literature from 1995 to 2014 was reviewed and focussed on assays performed with zebrafish larvae younger than 7 days post fertilization (dpf). The behavioural tests were scrutinized, and parameters describing the different experimental setups were defined. In the next step, we investigated if differences in the experimental parameters alter the outcome of the test. In order to test the comparability of behavioural assays, we analysed several studies using ethanol, valproate and pentylenetetrazole as model substances. Based on our findings, we provide recommendations which could help improve future behavioural studies performed with zebrafish larvae.
Collapse
Affiliation(s)
- J Legradi
- Institute for Environmental Studies, VU University, Amsterdam, The Netherlands.
| | - N el Abdellaoui
- Institute for Environmental Studies, VU University, Amsterdam, The Netherlands
| | - M van Pomeren
- Institute for Environmental Studies, VU University, Amsterdam, The Netherlands
| | - J Legler
- Institute for Environmental Studies, VU University, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Barbosa DJ, Capela JP, Feio-Azevedo R, Teixeira-Gomes A, Bastos MDL, Carvalho F. Mitochondria: key players in the neurotoxic effects of amphetamines. Arch Toxicol 2015; 89:1695-1725. [PMID: 25743372 DOI: 10.1007/s00204-015-1478-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/09/2015] [Indexed: 12/21/2022]
Abstract
Amphetamines are a class of psychotropic drugs with high abuse potential, as a result of their stimulant, euphoric, emphathogenic, entactogenic, and hallucinogenic properties. Although most amphetamines are synthetic drugs, of which methamphetamine, amphetamine, and 3,4-methylenedioxymethamphetamine ("ecstasy") represent well-recognized examples, the use of natural related compounds, namely cathinone and ephedrine, has been part of the history of humankind for thousands of years. Resulting from their amphiphilic nature, these drugs can easily cross the blood-brain barrier and elicit their well-known psychotropic effects. In the field of amphetamines' research, there is a general consensus that mitochondrial-dependent pathways can provide a major understanding concerning pathological processes underlying the neurotoxicity of these drugs. These events include alterations on tricarboxylic acid cycle's enzymes functioning, inhibition of mitochondrial electron transport chain's complexes, perturbations of mitochondrial clearance mechanisms, interference with mitochondrial dynamics, as well as oxidative modifications in mitochondrial macromolecules. Additionally, other studies indicate that amphetamines-induced neuronal toxicity is closely regulated by B cell lymphoma 2 superfamily of proteins with consequent activation of caspase-mediated downstream cell death pathway. Understanding the molecular mechanisms at mitochondrial level involved in amphetamines' neurotoxicity can help in defining target pathways or molecules mediating these effects, as well as in developing putative therapeutic approaches to prevent or treat the acute- or long-lasting neuropsychiatric complications seen in human abusers.
Collapse
Affiliation(s)
- Daniel José Barbosa
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.
| | - João Paulo Capela
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
- FP-ENAS (Unidade de Investigação UFP em energia, Ambiente e Saúde), CEBIMED (Centro de Estudos em Biomedicina), Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Rua 9 de Abril 349, 4249-004, Porto, Portugal
| | - Rita Feio-Azevedo
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Armanda Teixeira-Gomes
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
36
|
Bonneau N, Schmitz-Afonso I, Brunelle A, Touboul D, Champy P. Method development for quantification of the environmental neurotoxin annonacin in Rat plasma by UPLC-MS/MS and application to a pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 1004:46-52. [PMID: 26444335 DOI: 10.1016/j.jchromb.2015.09.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/21/2015] [Accepted: 09/24/2015] [Indexed: 11/30/2022]
Abstract
Annonacin is an environmental neurotoxin identified in the pulp of several fruits of the Annonaceae family (for example in Annona muricata, Asimina triloba), whose consumption was linked with the occurrence of sporadic atypical Parkinsonism with dementia. Pharmacokinetic parameters of this molecule are unknown. A method for its quantification in Rat plasma was developed, using its analogue annonacinone as an internal standard. Extraction from plasma was performed using ethylacetate with a good recovery. Quantification was performed by UPLC-MS/MS in SRM mode, based on the loss of the γ-methyl-γ-lactone (-112amu) from the sodium-cationized species [M+Na](+) of both annonacin and internal standard. The limit of quantification was 0.25ng/mL. Despite strong matrix effects, a good linearity was obtained over two distinct ranges 0.25-10ng/mL and 10-100ng/mL. The intra- and inter-day precisions (RSD) were lower than 10%, while accuracy was within ±10%. This method was applied to a pharmacokinetic study in the Rat. After oral administration of 10mg/kg annonacin, a Cmax of 7.9±1.5ng/mL was reached at Tmax 0.25h; T1/2 was 4.8±0.7h and apparent distribution volume was 387.9±64.6L. The bioavailability of annonacin was estimated to be 3.2±0.3% of the ingested dose.
Collapse
Affiliation(s)
- Natacha Bonneau
- Laboratoire de Pharmacognosie-Chimie des Substances Naturelles, UMR-BioCIS, CNRS, University Paris-Sud, Université Paris-Saclay, 5 rue Jean-Baptiste Clément, 9290, Châtenay-Malabry, France
| | - Isabelle Schmitz-Afonso
- Institut de Chimie des Substances Naturelles, CNRS-ICSN UPR2301, Université Paris-Sud, Labex LERMIT, avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Alain Brunelle
- Institut de Chimie des Substances Naturelles, CNRS-ICSN UPR2301, Université Paris-Sud, Labex LERMIT, avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - David Touboul
- Institut de Chimie des Substances Naturelles, CNRS-ICSN UPR2301, Université Paris-Sud, Labex LERMIT, avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Pierre Champy
- Laboratoire de Pharmacognosie-Chimie des Substances Naturelles, UMR-BioCIS, CNRS, University Paris-Sud, Université Paris-Saclay, 5 rue Jean-Baptiste Clément, 9290, Châtenay-Malabry, France.
| |
Collapse
|
37
|
Marthandan S, Priebe S, Groth M, Guthke R, Platzer M, Hemmerich P, Diekmann S. Hormetic effect of rotenone in primary human fibroblasts. Immun Ageing 2015; 12:11. [PMID: 26380578 PMCID: PMC4572608 DOI: 10.1186/s12979-015-0038-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/01/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Rotenone inhibits the electron transfer from complex I to ubiquinone, in this way interfering with the electron transport chain in mitochondria. This chain of events induces increased levels of intracellular reactive oxygen species, which in turn can contribute to acceleration of telomere shortening and induction of DNA damage, ultimately resulting in aging. In this study, we investigated the effect of rotenone treatment in human fibroblast strains. RESULTS For the first time we here describe that rotenone treatment induced a hormetic effect in human fibroblast strains. We identified a number of genes which were commonly differentially regulated due to low dose rotenone treatment in fibroblasts independent of their cell origin. However, these genes were not among the most strongly differentially regulated genes in the fibroblast strains on treatment with rotenone. Thus, if there is a common hormesis regulation, it is superimposed by cell strain specific individual responses. We found the rotenone induced differential regulation of pathways common between the two fibroblast strains, being weaker than the pathways individually regulated in the single fibroblast cell strains. Furthermore, within the common pathways different genes were responsible for this different regulation. Thus, rotenone induced hormesis was related to a weak pathway signal, superimposed by a stronger individual cellular response, a situation as found for the differentially expressed genes. CONCLUSION We found that the concept of hormesis also applies to in vitro aging of primary human fibroblasts. However, in depth analysis of the genes as well as the pathways differentially regulated due to rotenone treatment revealed cellular hormesis being related to weak signals which are superimposed by stronger individual cell-internal responses. This would explain that in general hormesis is a small effect. Our data indicate that the observed hormetic phenotype does not result from a specific strong well-defined gene or pathway regulation but from weak common cellular processes induced by low levels of reactive oxygen species. This conclusion also holds when comparing our results with those obtained for C. elegans in which the same low dose rotenone level induced a life span extending, thus hormetic effect.
Collapse
Affiliation(s)
- Shiva Marthandan
- />Leibniz-Institute for Age Research - Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Steffen Priebe
- />Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute e.V. (HKI), Jena, Germany
| | - Marco Groth
- />Leibniz-Institute for Age Research - Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Reinhard Guthke
- />Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute e.V. (HKI), Jena, Germany
| | - Matthias Platzer
- />Leibniz-Institute for Age Research - Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Peter Hemmerich
- />Leibniz-Institute for Age Research - Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Stephan Diekmann
- />Leibniz-Institute for Age Research - Fritz Lipmann Institute e.V. (FLI), Beutenbergstrasse 11, D-07745 Jena, Germany
| |
Collapse
|
38
|
Low Wang CC, Galinkin JL, Hiatt WR. Toxicity of a novel therapeutic agent targeting mitochondrial complex I. Clin Pharmacol Ther 2015; 98:551-9. [PMID: 26108785 DOI: 10.1002/cpt.178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/19/2015] [Indexed: 12/14/2022]
Abstract
R118 is an experimental compound that completed preclinical development as a potential medical therapy for the exercise limitation in peripheral artery disease. Animal studies established that R118 provided partial and reversible mitochondrial complex I inhibition with consequent increases in adenosine monophosphate (AMP) kinase activation in liver and skeletal muscle. After demonstration of improved exercise performance in a mouse model and safety in rodent and primate models, a phase I trial was performed in 24 subjects randomized to R118 vs. placebo (5:1) in escalating doses. Plasma lactic acid levels were transiently elevated in 20% of subjects at the lowest dose and in 100% of subjects using a different formulation at the highest dose, which was associated with hospitalization in all subjects and severe metabolic acidosis requiring prolonged intubation in two subjects. Thus, inhibition of mitochondrial complex I with R118 resulted in severe lactic acidosis, representing unacceptable toxicity from this mechanism of action.
Collapse
Affiliation(s)
- C C Low Wang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA.,CPC Clinical Research, Aurora, Colorado, USA
| | - J L Galinkin
- CPC Clinical Research, Aurora, Colorado, USA.,Department of Anesthesia, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - W R Hiatt
- CPC Clinical Research, Aurora, Colorado, USA.,Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
39
|
Castillo-Quan JI, Kinghorn KJ, Bjedov I. Genetics and pharmacology of longevity: the road to therapeutics for healthy aging. ADVANCES IN GENETICS 2015; 90:1-101. [PMID: 26296933 DOI: 10.1016/bs.adgen.2015.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging can be defined as the progressive decline in tissue and organismal function and the ability to respond to stress that occurs in association with homeostatic failure and the accumulation of molecular damage. Aging is the biggest risk factor for human disease and results in a wide range of aging pathologies. Although we do not completely understand the underlying molecular basis that drives the aging process, we have gained exceptional insights into the plasticity of life span and healthspan from the use of model organisms such as the worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Single-gene mutations in key cellular pathways that regulate environmental sensing, and the response to stress, have been identified that prolong life span across evolution from yeast to mammals. These genetic manipulations also correlate with a delay in the onset of tissue and organismal dysfunction. While the molecular genetics of aging will remain a prosperous and attractive area of research in biogerontology, we are moving towards an era defined by the search for therapeutic drugs that promote healthy aging. Translational biogerontology will require incorporation of both therapeutic and pharmacological concepts. The use of model organisms will remain central to the quest for drug discovery, but as we uncover molecular processes regulated by repurposed drugs and polypharmacy, studies of pharmacodynamics and pharmacokinetics, drug-drug interactions, drug toxicity, and therapeutic index will slowly become more prevalent in aging research. As we move from genetics to pharmacology and therapeutics, studies will not only require demonstration of life span extension and an underlying molecular mechanism, but also the translational relevance for human health and disease prevention.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J Kinghorn
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Ivana Bjedov
- Cancer Institute, University College London, London, UK
| |
Collapse
|
40
|
Blesa J, Trigo-Damas I, Quiroga-Varela A, Jackson-Lewis VR. Oxidative stress and Parkinson's disease. Front Neuroanat 2015. [PMID: 26217195 PMCID: PMC4495335 DOI: 10.3389/fnana.2015.00091] [Citation(s) in RCA: 548] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parkinson disease (PD) is a chronic, progressive neurological disease that is associated with a loss of dopaminergic neurons in the substantia nigra pars compacta of the brain. The molecular mechanisms underlying the loss of these neurons still remain elusive. Oxidative stress is thought to play an important role in dopaminergic neurotoxicity. Complex I deficiencies of the respiratory chain account for the majority of unfavorable neuronal degeneration in PD. Environmental factors, such as neurotoxins, pesticides, insecticides, dopamine (DA) itself, and genetic mutations in PD-associated proteins contribute to mitochondrial dysfunction which precedes reactive oxygen species formation. In this mini review, we give an update of the classical pathways involving these mechanisms of neurodegeneration, the biochemical and molecular events that mediate or regulate DA neuronal vulnerability, and the role of PD-related gene products in modulating cellular responses to oxidative stress in the course of the neurodegenerative process.
Collapse
Affiliation(s)
- Javier Blesa
- Centro Integral de Neurociencias A.C., HM Puerta del Sur, Hospitales de Madrid, Móstoles and Medical School, CEU San Pablo University, Madrid Spain
| | - Ines Trigo-Damas
- Centro Integral de Neurociencias A.C., HM Puerta del Sur, Hospitales de Madrid, Móstoles and Medical School, CEU San Pablo University, Madrid Spain
| | - Anna Quiroga-Varela
- Department of Medicine, Clinica Neurologica, Ospedale Santa Maria della Misericordia - Università di Perugia, Perugia Italy
| | | |
Collapse
|
41
|
Parameshwaran K, Irwin MH, Steliou K, Suppiramaniam V, Pinkert CA. Antioxidant-Mediated Reversal of Oxidative Damage in Mouse Modeling of Complex I Inhibition. Drug Dev Res 2015; 76:72-81. [DOI: 10.1002/ddr.21242] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/14/2014] [Indexed: 12/17/2022]
Affiliation(s)
| | - Michael H. Irwin
- Department of Pathobiology, College of Veterinary Medicine; Auburn University; Auburn AL USA
| | - Kosta Steliou
- PhenoMatriX, Inc., Boston, MA, and Cancer Research Center; Boston University School of Medicine; Boston MA USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy; Auburn University; Auburn AL USA
| | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary Medicine; Auburn University; Auburn AL USA
- Department of Biological Sciences; University of Alabama; Tuscaloosa AL USA
| |
Collapse
|
42
|
Skemiene K, Liobikas J, Borutaite V. Anthocyanins as substrates for mitochondrial complex I - protective effect against heart ischemic injury. FEBS J 2015; 282:963-71. [PMID: 25586661 DOI: 10.1111/febs.13195] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 12/21/2014] [Accepted: 01/10/2015] [Indexed: 12/12/2022]
Abstract
Anthocyanins, a subclass of flavonoids, are known to protect against myocardial ischemia; however, little is known about their direct, acute effects on mitochondria injured by the ischemic insult. In this study, the effects of delphinidin 3-O-glucoside (Dp3G), cyanidin 3-O-glucoside (Cy3G) and pelargonidin 3-O-glucoside (Pg3G) on the activity of complex I of the mitochondrial respiratory chain were studied in mitochondria isolated from normal rat hearts and rat hearts subjected to ischemia for 45 min. Cy3G and Dp3G increased the activity of complex I, measured in the presence or absence of coenzyme Q1 (CoQ1 ), in ischemia-damaged mitochondria, whereas in nonischemic mitochondria the effect was observed only in the absence of CoQ1 . Dp3G and Cy3G but not Pg3G increased state 3 respiration and ATP synthesis with NADH-dependent substrates in mitochondria after ischemia. The results suggest that certain anthocyanins can act as electron acceptors at complex I, and bypass ischemia-induced inhibition, resulting in increased ATP production after ischemia. This study provides new information on a possible role of certain anthocyanins in the regulation of energy metabolism in mammalian cells.
Collapse
Affiliation(s)
- Kristina Skemiene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | |
Collapse
|
43
|
Long-term and low-dose malathion exposure causes cognitive impairment in adult mice: evidence of hippocampal mitochondrial dysfunction, astrogliosis and apoptotic events. Arch Toxicol 2015; 90:647-60. [DOI: 10.1007/s00204-015-1466-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022]
|
44
|
Okatsu K, Kimura M, Oka T, Tanaka K, Matsuda N. Unconventional PINK1 localization to the outer membrane of depolarized mitochondria drives Parkin recruitment. J Cell Sci 2015; 128:964-78. [PMID: 25609704 PMCID: PMC4342580 DOI: 10.1242/jcs.161000] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Dysfunction of PTEN-induced putative kinase 1 (PINK1), a Ser/Thr kinase with an N-terminal mitochondrial-targeting sequence (MTS), causes familial recessive parkinsonism. Reduction of the mitochondrial membrane potential limits MTS-mediated matrix import and promotes PINK1 accumulation on the outer mitochondrial membrane (OMM) of depolarized mitochondria. PINK1 then undergoes autophosphorylation and phosphorylates ubiquitin and Parkin, a cytosolic ubiquitin ligase, for clearance of damaged mitochondria. The molecular basis for PINK1 localization on the OMM of depolarized mitochondria rather than release to the cytosol is poorly understood. Here, we disentangle the PINK1 localization mechanism using deletion mutants and a newly established constitutively active PINK1 mutant. Disruption of the MTS through N-terminal insertion of aspartic acid residues results in OMM localization of PINK1 in energized mitochondria. Unexpectedly, the MTS and putative transmembrane domain (TMD) are dispensable for OMM localization, whereas mitochondrial translocase Tom40 (also known as TOMM40) and an alternative mitochondrial localization signal that resides between the MTS and TMD are required. PINK1 utilizes a mitochondrial localization mechanism that is distinct from that of conventional MTS proteins and that presumably functions in conjunction with the Tom complex in OMM localization when the conventional N-terminal MTS is inhibited.
Collapse
Affiliation(s)
- Kei Okatsu
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Mayumi Kimura
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Toshihiko Oka
- Department of Life Science, College of Science, Rikkyo University, Nishi-Ikebukuro, Tokyo 171-8501, Japan
| | - Keiji Tanaka
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Noriyuki Matsuda
- Protein Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
45
|
Gehrke S, Wu Z, Klinkenberg M, Sun Y, Auburger G, Guo S, Lu B. PINK1 and Parkin control localized translation of respiratory chain component mRNAs on mitochondria outer membrane. Cell Metab 2015; 21:95-108. [PMID: 25565208 PMCID: PMC4455944 DOI: 10.1016/j.cmet.2014.12.007] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/14/2014] [Accepted: 12/13/2014] [Indexed: 11/15/2022]
Abstract
Mitochondria play essential roles in many aspects of biology, and their dysfunction has been linked to diverse diseases. Central to mitochondrial function is oxidative phosphorylation (OXPHOS), accomplished by respiratory chain complexes (RCCs) encoded by nuclear and mitochondrial genomes. How RCC biogenesis is regulated in metazoans is poorly understood. Here we show that Parkinson's disease (PD)-associated genes PINK1 and Parkin direct localized translation of certain nuclear-encoded RCC (nRCC) mRNAs. Translationally repressed nRCC mRNAs are localized in a PINK1/Tom20-dependent manner to mitochondrial outer membrane, where they are derepressed and activated by PINK1/Parkin through displacement of translation repressors, including Pumilio and Glorund/hnRNP-F, a Parkin substrate, and enhanced binding of activators such as eIF4G. Inhibiting the translation repressors rescued nRCC mRNA translation and neuromuscular-degeneration phenotypes of PINK1 mutant, whereas inhibiting eIF4G had opposite effects. Our results reveal previously unknown functions of PINK1/Parkin in RNA metabolism and suggest new approaches to mitochondrial restoration and disease intervention.
Collapse
Affiliation(s)
- Stephan Gehrke
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zhihao Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Klinkenberg
- Experimental Neurology, Goethe University Medical School, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Yaping Sun
- Department of Bioengineering and Therapeutic Sciences, Programs in Biological Sciences and Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Georg Auburger
- Experimental Neurology, Goethe University Medical School, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, Programs in Biological Sciences and Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
46
|
Bruch J, Xu H, De Andrade A, Höglinger G. Mitochondrial complex 1 inhibition increases 4-repeat isoform tau by SRSF2 upregulation. PLoS One 2014; 9:e113070. [PMID: 25402454 PMCID: PMC4234644 DOI: 10.1371/journal.pone.0113070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/23/2014] [Indexed: 12/16/2022] Open
Abstract
Progressive Supranuclear Palsy (PSP) is a neurodegenerative disorder characterised by intracellular aggregation of the microtubule-associated protein tau. The tau protein exists in 6 predominant isoforms. Depending on alternative splicing of exon 10, three of these isoforms have four microtubule-binding repeat domains (4R), whilst the others only have three (3R). In PSP there is an excess of the 4R tau isoforms, which are thought to contribute significantly to the pathological process. The cause of this 4R increase is so far unknown. Several lines of evidence link mitochondrial complex I inhibition to the pathogenesis of PSP. We demonstrate here for the first time that annonacin and MPP+, two prototypical mitochondrial complex I inhibitors, increase the 4R isoforms of tau in human neurons. We show that the splicing factor SRSF2 is necessary to increase 4R tau with complex I inhibition. We also found SRSF2, as well as another tau splicing factor, TRA2B, to be increased in brains of PSP patients. Thereby, we provide new evidence that mitochondrial complex I inhibition may contribute as an upstream event to the pathogenesis of PSP and suggest that splicing factors may represent an attractive therapeutic target to intervene in the disease process.
Collapse
Affiliation(s)
- Julius Bruch
- Department of Translational Neurodegeneration, German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
| | - Hong Xu
- Department of Translational Neurodegeneration, German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
| | - Anderson De Andrade
- Department of Translational Neurodegeneration, German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Günter Höglinger
- Department of Translational Neurodegeneration, German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
47
|
Affiliation(s)
- Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94321, USA.
| |
Collapse
|
48
|
The neurotoxicity of 5-S-cysteinyldopamine is mediated by the early activation of ERK1/2 followed by the subsequent activation of ASK1/JNK1/2 pro-apoptotic signalling. Biochem J 2014; 463:41-52. [DOI: 10.1042/bj20131519] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
5-S-cysteinyldopamine is an endogenous neurotoxin with relevance to Parkinson's disease. The present study shows for the first time that the endogenous formation of 5-S-cysteinyldopamine in the Parkinsonian brain may be causally related to nigrostriatal tract degeneration.
Collapse
|
49
|
Batandier C, Poulet L, Hininger I, Couturier K, Fontaine E, Roussel AM, Canini F. Acute stress delays brain mitochondrial permeability transition pore opening. J Neurochem 2014; 131:314-22. [PMID: 24989320 DOI: 10.1111/jnc.12811] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/28/2014] [Accepted: 06/28/2014] [Indexed: 12/15/2022]
Abstract
Since emotional stress elicits brain activation, mitochondria should be a key component of stressed brain response. However, few studies have focused on mitochondria functioning in these conditions. In this work, we aimed to determine the effects of an acute restraint stress on rat brain mitochondrial functions, with a focus on permeability transition pore (PTP) functioning. Rats were divided into two groups, submitted or not to an acute 30-min restraint stress (Stress, S-group, vs. Control, C-group). Brain was removed immediately after stress. Mitochondrial respiration and enzymatic activities (complex I, complex II, hexokinase) were measured. Changes in PTP opening were assessed by the Ca(2+) retention capacity. Cell signaling pathways relevant to the coupling between mitochondria and cell function (adenosine monophosphate-activated protein kinase, phosphatidylinositol 3-kinase, glycogen synthase kinase 3 beta, MAPK, and cGMP/NO) were measured. The effect of glucocorticoids was also assessed in vitro. Stress delayed (43%) the opening of PTP and resulted in a mild inhibition of complex I respiratory chain. This inhibition was associated with significant stress-induced changes in adenosine monophosphate-activated protein kinase signaling pathway without changes in brain cGMP level. In contrast, glucocorticoids did not modify PTP opening. These data suggest a rapid adaptive mechanism of brain mitochondria in stressed conditions, with a special focus on PTP regulation. In a rat model of acute restraint stress, we observed substantial changes in brain mitochondria functioning. Stress significantly (i) delays (43%) the opening of permeability transition pore (PTP) by the calcium (Ca(2+) ), its main inductor and (ii) results in an inhibition of complex I in electron transport chain associated with change in AMPK signaling pathway. These data suggest an adaptive mechanism of brain mitochondria in stressed condition, with a special focus on PTP regulation.
Collapse
Affiliation(s)
- Cécile Batandier
- Laboratoire de Bioénergétique Fondamentale et Appliquée, Université Joseph Fourier, Grenoble, France; U1055 - INSERM, Grenoble, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Koyano F, Okatsu K, Kosako H, Tamura Y, Go E, Kimura M, Kimura Y, Tsuchiya H, Yoshihara H, Hirokawa T, Endo T, Fon EA, Trempe JF, Saeki Y, Tanaka K, Matsuda N. Ubiquitin is phosphorylated by PINK1 to activate parkin. Nature 2014; 510:162-6. [DOI: 10.1038/nature13392] [Citation(s) in RCA: 1001] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/23/2014] [Indexed: 12/28/2022]
|