51
|
Reduced Function of the Glutathione S-Transferase S1 Suppresses Behavioral Hyperexcitability in Drosophila Expressing Mutant Voltage-Gated Sodium Channels. G3-GENES GENOMES GENETICS 2020; 10:1327-1340. [PMID: 32054635 PMCID: PMC7144092 DOI: 10.1534/g3.119.401025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Voltage-gated sodium (Nav) channels play a central role in the generation and propagation of action potentials in excitable cells such as neurons and muscles. To determine how the phenotypes of Nav-channel mutants are affected by other genes, we performed a forward genetic screen for dominant modifiers of the seizure-prone, gain-of-function Drosophila melanogaster Nav-channel mutant, paraShu. Our analyses using chromosome deficiencies, gene-specific RNA interference, and single-gene mutants revealed that a null allele of glutathione S-transferase S1 (GstS1) dominantly suppresses paraShu phenotypes. Reduced GstS1 function also suppressed phenotypes of other seizure-prone Nav-channel mutants, paraGEFS+ and parabss. Notably, paraShu mutants expressed 50% less GstS1 than wild-type flies, further supporting the notion that paraShu and GstS1 interact functionally. Introduction of a loss-of-function GstS1 mutation into a paraShu background led to up- and down-regulation of various genes, with those encoding cytochrome P450 (CYP) enzymes most significantly over-represented in this group. Because GstS1 is a fly ortholog of mammalian hematopoietic prostaglandin D synthase, and in mammals CYPs are involved in the oxygenation of polyunsaturated fatty acids including prostaglandins, our results raise the intriguing possibility that bioactive lipids play a role in GstS1-mediated suppression of paraShu phenotypes.
Collapse
|
52
|
Manzoor R, Rasool A, Ahmed M, Kaleem U, Duru LN, Ma H, Deng Y. Synergistic Neuroprotective Effect of Endogenously-Produced Hydroxytyrosol and Synaptic Vesicle Proteins on Pheochromocytoma Cell Line Against Salsolinol. Molecules 2020; 25:E1715. [PMID: 32276517 PMCID: PMC7181248 DOI: 10.3390/molecules25071715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 01/29/2023] Open
Abstract
Oxidative stress triggers a lethal cascade, leading to Parkinson's disease by causing degeneration of dopaminergic neurons. In this study, eight antioxidants were screened for their neuroprotective effect on PC12 cells (pheochromocytoma cell line) under oxidative stress induced by salsolinol (OSibS). Hydroxytyrosol was found to be the strongest neuroprotective agent; it improved viability of PC12 cells by up to 81.69% under OSibS. Afterward, two synaptic vesicle proteins, synapsin-1 and septin-5, were screened for their neuroprotective role; the overexpression of synapsin-1 and the downregulation of septin-5 separately improved the viability of PC12 cells by up to 71.17% and 67.00%, respectively, compared to PC12 cells only treated with salsolinol (PoTwS) under OSibS. Subsequently, the PC12+syn++sep- cell line was constructed and pretreated with 100 µM hydroxytyrosol, which improved its cell viability by up to 99.03% and led to 14.71- and 6.37-fold reductions in the levels of MDA and H2O2, respectively, and 6.8-, 12.97-, 10.57-, and 7.57-fold increases in the activity of catalase, glutathione reductase, superoxide dismutase, and glutathione peroxidase, respectively, compared to PoTwS under OSibS. Finally, alcohol dehydrogenase-6 from Saccharomyces cerevisiae was expressed in PC12+syn++sep- cells to convert 3,4-dihydroxyphenylacetaldehyde (an endogenous neurotoxin) into hydroxytyrosol. The PC12+syn++sep-+ADH6+ cell line also led to 22.38- and 12.33-fold decreases in the production of MDA and H2O2, respectively, and 7.15-, 13.93-, 12.08-, and 8.11-fold improvements in the activity of catalase, glutathione reductase, superoxide dismutase, and glutathione peroxidase, respectively, compared to PoTwS under OSibS. Herein, we report the endogenous production of a powerful antioxidant, hydroxytyrosol, from 3,4-dihydroxyphenylacetaldehyde, and evaluate its synergistic neuroprotective effect, along with synapsin-1 and septin-5, on PC12 cells under OSibS.
Collapse
Affiliation(s)
- Robina Manzoor
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (R.M.); (L.N.D.); (H.M.)
| | - Aamir Rasool
- Institute for Synthetic Biosystem, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China;
- Institute of Biochemistry, University of Balochistan, Quetta 87300, Pakistan
| | - Maqbool Ahmed
- Department of Tuberculosis, Bolan University of Medical and Health Sciences, Quetta 87300, Pakistan;
| | - Ullah Kaleem
- Department of Microbiology, University of Balochistan, Quetta 87300, Pakistan;
| | - Lucienne Nneoma Duru
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (R.M.); (L.N.D.); (H.M.)
| | - Hong Ma
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (R.M.); (L.N.D.); (H.M.)
| | - Yulin Deng
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (R.M.); (L.N.D.); (H.M.)
| |
Collapse
|
53
|
Park Y, Ku L, Lim JW, Kim H. Docosahexaenoic acid inhibits zymogen activation by suppressing vacuolar ATPase activation in cerulein-stimulated pancreatic acinar cells. GENES AND NUTRITION 2020; 15:6. [PMID: 32293245 PMCID: PMC7092610 DOI: 10.1186/s12263-020-00664-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 03/05/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND The premature activation of digestive enzyme zymogens within pancreatic acinar cells is an important early feature of acute pancreatitis. Supraphysiological concentrations of cholecystokinin (CCK) cause intrapancreatic zymogen activation and acute pancreatitis. Stimulation of vacuolar ATPase (vATPase) activity is required for zymogen activation in pancreatic acinar cells. Parkin, a multiprotein E3 ubiquitin ligase complex, promotes vATPase ubiquitination and degradation, which inhibits vATPase activity. Docosahexaenoic acid (DHA), an omega-3 fatty acid, exerts anti-inflammatory effects. It is reported to bind to G-protein coupled receptor 120 (GPR120) and GPR40. DHA induces the degradation of certain proteins by activating ubiquitin-proteasome system in various cells. This study aimed to investigate whether DHA induces Parkin and inhibits vATPase activity, resulting in zymogen inactivation in pancreatic acinar AR42J cells stimulated with cerulein, a CCK analog. RESULTS Cerulein induced the translocation of the cytosolic V1 domain (E subunit) of vATPase to the membrane, which indicated vATPase activation, and zymogen activation in AR42J cells. DHA suppressed the association of the vATPase with membranes, and zymogen activation (increased trypsin activity and amylase release) induced by cerulein. Pretreatment with a GPR120 antagonist AH-7614, a GPR40 antagonist DC260126, or an ubiquitination inhibitor PYR-41 reduced the effect of DHA on cerulein-induced zymogen activation. Treatment with PYR-41 reversed the DHA-induced decrease in vATPase activation in cerulein-treated cells. Furthermore, DHA increased the level of Parkin in membranes of cerulein-treated cells. CONCLUSIONS DHA upregulates Parkin which inhibits vATPase-mediated zymogen activation, via GPR120 and GPR40, in cerulein-stimulated pancreatic acinar cells.
Collapse
Affiliation(s)
- Yeeun Park
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Leeyeon Ku
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Joo Weon Lim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Hyeyoung Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
54
|
Matsuda N, Yamano K. Two sides of a coin: Physiological significance and molecular mechanisms for damage-induced mitochondrial localization of PINK1 and Parkin. Neurosci Res 2020; 159:16-24. [PMID: 32201358 DOI: 10.1016/j.neures.2020.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022]
Abstract
In 1998, PARKIN was reported as a causal gene for hereditary recessive Parkinsonism by Kitada, Mizuno, Hattori, and Shimizu et al. Later in 2004, PINK1 was also reported as a causal gene for hereditary recessive Parkinsonism by Valente, Auburger, and Wood et al. Although many unsolved mysteries still remain, our knowledge of PINK1 and Parkin function has increased dramatically since then. Despite a number of milestone studies that advanced the PINK1 and Parkin research field, a critical turning point was undoubtedly the determination that their genuine subcellular localization was on depolarized mitochondria. In this review, we outline the key studies that have contributed to our current model for mitochondrial localization of PINK1 and Parkin. Interestingly, like two sides of a coin, our attempts to elucidate the mechanisms underlying the localization of PINK1 and Parkin were inextricably tied to the identification of the PINK1 substrate and molecular dissection of the Parkin activation mechanism.
Collapse
Affiliation(s)
- Noriyuki Matsuda
- The Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Room N-202, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| | - Koji Yamano
- The Ubiquitin Project, Tokyo Metropolitan Institute of Medical Science, Room N-202, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| |
Collapse
|
55
|
Calogero AM, Mazzetti S, Pezzoli G, Cappelletti G. Neuronal microtubules and proteins linked to Parkinson's disease: a relevant interaction? Biol Chem 2020; 400:1099-1112. [PMID: 31256059 DOI: 10.1515/hsz-2019-0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022]
Abstract
Neuronal microtubules are key determinants of cell morphology, differentiation, migration and polarity, and contribute to intracellular trafficking along axons and dendrites. Microtubules are strictly regulated and alterations in their dynamics can lead to catastrophic effects in the neuron. Indeed, the importance of the microtubule cytoskeleton in many human diseases is emerging. Remarkably, a growing body of evidence indicates that microtubule defects could be linked to Parkinson's disease pathogenesis. Only a few of the causes of the progressive neuronal loss underlying this disorder have been identified. They include gene mutations and toxin exposure, but the trigger leading to neurodegeneration is still unknown. In this scenario, the evidence showing that mutated proteins in Parkinson's disease are involved in the regulation of the microtubule cytoskeleton is intriguing. Here, we focus on α-Synuclein, Parkin and Leucine-rich repeat kinase 2 (LRRK2), the three main proteins linked to the familial forms of the disease. The aim is to dissect their interaction with tubulin and microtubules in both physiological and pathological conditions, in which these proteins are overexpressed, mutated or absent. We highlight the relevance of such an interaction and suggest that these proteins could trigger neurodegeneration via defective regulation of the microtubule cytoskeleton.
Collapse
Affiliation(s)
- Alessandra M Calogero
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy
| | - Samanta Mazzetti
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy.,Fondazione Grigioni per il Morbo di Parkinson, via Zuretti 35, I-20135 Milan, Italy
| | - Gianni Pezzoli
- Fondazione Grigioni per il Morbo di Parkinson, via Zuretti 35, I-20135 Milan, Italy.,Parkinson Institute, ASST "G.Pini-CTO", via Bignami 1, I-20133 Milan, Italy
| | - Graziella Cappelletti
- Department of Biosciences, Università degli Studi di Milano, via Celoria 26, I-20133 Milan, Italy.,Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, via Balzaretti, I-20133 Milan, Italy
| |
Collapse
|
56
|
Villanueva-Paz M, Povea-Cabello S, Villalón-García I, Álvarez-Córdoba M, Suárez-Rivero JM, Talaverón-Rey M, Jackson S, Falcón-Moya R, Rodríguez-Moreno A, Sánchez-Alcázar JA. Parkin-mediated mitophagy and autophagy flux disruption in cellular models of MERRF syndrome. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165726. [PMID: 32061767 DOI: 10.1016/j.bbadis.2020.165726] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 01/16/2020] [Accepted: 02/10/2020] [Indexed: 12/27/2022]
Abstract
Mitochondrial diseases are considered rare genetic disorders characterized by defects in oxidative phosphorylation (OXPHOS). They can be provoked by mutations in nuclear DNA (nDNA) or mitochondrial DNA (mtDNA). MERRF (Myoclonic Epilepsy with Ragged-Red Fibers) syndrome is one of the most frequent mitochondrial diseases, principally caused by the m.8344A>G mutation in mtDNA, which affects the translation of all mtDNA-encoded proteins and therefore impairs mitochondrial function. In the present work, we evaluated autophagy and mitophagy flux in transmitochondrial cybrids and fibroblasts derived from a MERRF patient, reporting that Parkin-mediated mitophagy is increased in MERRF cell cultures. Our results suggest that supplementation with coenzyme Q10 (CoQ), a component of the electron transport chain (ETC) and lipid antioxidant, prevents Parkin translocation to the mitochondria. In addition, CoQ acts as an enhancer of autophagy and mitophagy flux, which partially improves cell pathophysiology. The significance of Parkin-mediated mitophagy in cell survival was evaluated by silencing the expression of Parkin in MERRF cybrids. Our results show that mitophagy acts as a cell survival mechanism in mutant cells. To confirm these results in one of the main affected cell types in MERRF syndrome, mutant induced neurons (iNs) were generated by direct reprogramming of patients-derived skin fibroblasts. The treatment of MERRF iNs with Guttaquinon CoQ10 (GuttaQ), a water-soluble derivative of CoQ, revealed a significant improvement in cell bioenergetics. These results indicate that iNs, along with fibroblasts and cybrids, can be utilized as reliable cellular models to shed light on disease pathomechanisms as well as for drug screening.
Collapse
Affiliation(s)
- Marina Villanueva-Paz
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Juan M Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain
| | - Sandra Jackson
- Department of Neurology, Uniklinikum C. G. Carus, Dresden, Germany
| | - Rafael Falcón-Moya
- Laboratorio de Neurociencia Celular y Plasticidad, Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla 41013, Spain
| | - Antonio Rodríguez-Moreno
- Laboratorio de Neurociencia Celular y Plasticidad, Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla 41013, Spain
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), Spain; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
57
|
Imai Y. PINK1-Parkin signaling in Parkinson's disease: Lessons from Drosophila. Neurosci Res 2020; 159:40-46. [PMID: 32035987 DOI: 10.1016/j.neures.2020.01.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 01/30/2020] [Indexed: 12/30/2022]
Abstract
The mitochondrial protein kinase PINK1 activates Parkin ubiquitin ligase by phosphorylating Parkin and ubiquitin, which are required for mitochondrial maintenance in dopaminergic (DA) neurons whose degeneration leads to the development of Parkinson's disease (PD). Loss of PINK1 and Parkin leads to mitochondrial degeneration and abnormal wing posture in Drosophila. Modifier screening using the Drosophila wing phenotype showed that the inactivation of Miro, a mitochondrial adaptor protein, suppresses the phenotype caused by mitochondrial degeneration. When activated by PINK1, Parkin suppresses mitochondrial transport by reducing Miro levels in Drosophila DA neurons. In human DA neurons, PINK1-Parkin signaling also regulates axonal mitochondrial re-distribution in response to reduced mitochondrial membrane potential, which is impaired in the DA neurons of patients with PINK1 and Parkin mutations. Phospho-ubiquitin signals amplified by PINK1 and Parkin are stronger in DA neurons than other neurons, suggesting that PINK1-Parkin signaling is particularly important for DA neuron activity. Moreover, the recently identified PD-associated protein CHCHD2 may ensure proper electron transfer during mitochondrial respiration. The genetic interaction between PINK1/Parkin and CHCHD2 in Drosophila indicates that they are not directly associated and CHCHD2-linked PD exhibits a very different pathology to PINK1/Parkin PD. I suggest a complex pathogenesis for mitochondrial dysregulation in PD.
Collapse
Affiliation(s)
- Yuzuru Imai
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.
| |
Collapse
|
58
|
Rai SN, Singh P. Advancement in the modelling and therapeutics of Parkinson's disease. J Chem Neuroanat 2020; 104:101752. [PMID: 31996329 DOI: 10.1016/j.jchemneu.2020.101752] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/19/2020] [Accepted: 01/20/2020] [Indexed: 02/08/2023]
Abstract
Since the discovery of L-dopa in the middle of the 20th century (1960s), there is not any neuroprotective therapy available although significant development has been made in the treatment of symptomatic Parkinson's disease (PD). Neurological disorders like PD can be modelled in animals so as to recapitulates most of the symptoms seen in PD patients. In aging population, PD is the second most common neurodegenerative disease after Alzheimer's disease, even though significant outcomes have been achieved in PD research yet it still is a mystery to solve the treatments for PD. In the last two decades, PD models have provided enhanced precision into the understanding of the process of PD disease, its etiology, pathology, and molecular mechanisms behind it. Furthermore, at the same time as cellular models have helped to recognize specific events, animal models, both toxic and genetic, have replicated almost all of the hallmarks of PD and are very helpful for testing and finding new strategies for neuroprotection. Recently, in both classical and newer models, major advances have been done in the modelling of supplementary PD features have come into the light. In this review, we have try to provide an updated summary of the characteristics of these models related to in vitro and in vivo models, animal models for PD, stem cell model for PD, newer 3D model as well as the strengths and limitations of these most popular PD models.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Department of Zoology, Mahila Maha Vidhyalaya, Institute of Science, Banaras Hindu University, Varanasi, India.
| | - Payal Singh
- Department of Zoology, Mahila Maha Vidhyalaya, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
59
|
Bayne AN, Trempe JF. Mechanisms of PINK1, ubiquitin and Parkin interactions in mitochondrial quality control and beyond. Cell Mol Life Sci 2019; 76:4589-4611. [PMID: 31254044 PMCID: PMC11105328 DOI: 10.1007/s00018-019-03203-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is a degenerative movement disorder resulting from the loss of specific neuron types in the midbrain. Early environmental and pathophysiological studies implicated mitochondrial damage and protein aggregation as the main causes of PD. These findings are now vindicated by the characterization of more than 20 genes implicated in rare familial forms of the disease. In particular, two proteins encoded by the Parkin and PINK1 genes, whose mutations cause early-onset autosomal recessive PD, function together in a mitochondrial quality control pathway. In this review, we will describe recent development in our understanding of their mechanisms of action, structure, and function. We explain how PINK1 acts as a mitochondrial damage sensor via the regulated proteolysis of its N-terminus and the phosphorylation of ubiquitin tethered to outer mitochondrial membrane proteins. In turn, phospho-ubiquitin recruits and activates Parkin via conformational changes that increase its ubiquitin ligase activity. We then describe how the formation of polyubiquitin chains on mitochondria triggers the recruitment of the autophagy machinery or the formation of mitochondria-derived vesicles. Finally, we discuss the evidence for the involvement of these mechanisms in physiological processes such as immunity and inflammation, as well as the links to other PD genes.
Collapse
Affiliation(s)
- Andrew N Bayne
- Department of Pharmacology and Therapeutics and Centre for Structural Biology, McGill University, 3655 Prom Sir William Osler, Montreal, QC, H3G 1Y6, Canada
| | - Jean-François Trempe
- Department of Pharmacology and Therapeutics and Centre for Structural Biology, McGill University, 3655 Prom Sir William Osler, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
60
|
Post-translational modification of Parkin and its research progress in cancer. Cancer Commun (Lond) 2019; 39:77. [PMID: 31753025 PMCID: PMC6873554 DOI: 10.1186/s40880-019-0421-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022] Open
Abstract
Clinical practice has shown that Parkin is the major causative gene found in an autosomal recessive juvenile parkinsonism (AR-JP) via Parkin mutations and that the Parkin protein is the core expression product of the Parkin gene, which itself belongs to an E3 ubiquitin ligase. Since the discovery of the Parkin gene in the late 1990s, researchers in many countries have begun extensive research on this gene and found that in addition to AR-JP, the Parkin gene is associated with many diseases, including type 2 diabetes, leprosy, Alzheimer’s, autism, and cancer. Recent studies have found that the loss or dysfunction of Parkin has a certain relationship with tumorigenesis. In general, the Parkin gene, a well-established tumor suppressor, is deficient and mutated in a variety of malignancies. Parkin overexpression inhibits tumor cell growth and promotes apoptosis. However, the functions of Parkin in tumorigenesis and its regulatory mechanisms are still not fully understood. This article describes the structure, functions, and post-translational modifications of Parkin, and summarizes the recent advances in the tumor suppressive function of Parkin and its underlying mechanisms.
Collapse
|
61
|
Parkin truncating variants result in a loss-of-function phenotype. Sci Rep 2019; 9:16150. [PMID: 31695088 PMCID: PMC6834586 DOI: 10.1038/s41598-019-52534-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 10/16/2019] [Indexed: 11/08/2022] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder. Most cases of PD are sporadic, while 5-10% have a known genetic basis. Variants in the PARK2 gene are the most frequent cause of autosomal recessive juvenile-onset PD. PARK2 encodes parkin, a multi-domain protein that functions as an ubiquitin E3 ligase. Numerous variants spanning all parkin domains have been identified, although the pathogenic relevance for several of those remains unclear. In this study, we aimed to functionally characterize two truncating parkin variants: N52Mfs*29, which is highly prevalent in the Portuguese and Spanish populations, and L358Rfs*77, recently identified in the Portuguese population. Our results indicate that both variants are prematurely degraded by the proteasome, even though proteins levels are still moderate. We also showed that they are aggregation-prone and lead to mislocalized parkin. Interestingly, the L358Rfs*77 variant is mislocalized to the nucleus, which was never reported for parkin variants. While N52Mfs*29 impaired self-ubiquitination activity, the L358Rfs*77 variant seemed to retain it. Both variants, however, fail to ubiquitinate p62 substrate and did not relocalize to depolarized mitochondria. Therefore, we conclude that parkin truncating variants cause loss of parkin function, thus showing their causative role in PD pathogenesis.
Collapse
|
62
|
Lei R, Shen J, Zhang S, Liu A, Chen X, Wang Y, Sun J, Dai S, Xu J. Inactivating the ubiquitin ligase Parkin suppresses cell proliferation and induces apoptosis in human keloids. J Cell Physiol 2019; 234:16601-16608. [PMID: 30784061 DOI: 10.1002/jcp.28332] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 01/24/2023]
Abstract
Keloids are a common type of pathological skin healing, characterized by the destruction of the vascular network. Thus, keloids often exhibit anoxic conditions. Hypoxia-inducible factor-1α (HIF-1α) is a core factor that mediates hypoxia stress responses and allows the cells to adapt to low-oxygen conditions. In the current study, we identified that Parkin acted as an E3 ubiquitin ligase, contributing to the degradation of HIF-1α in keloid fibroblasts (KFs). Silencing of Parkin in KFs upregulated HIF-1α expression and prolonged its protein half-life. Furthermore, Parkin influenced transforming growth factor β (TGF-β)/Smad signaling by targeting HIF-1α. Under hypoxic conditions, silencing Parkin enhanced KF proliferation and inhibited apoptosis through the TGF-β/Smad signaling pathway. Notably, metformin, an antidiabetic drug, could significantly induce Parkin expression and enhance the interaction between Parkin and HIF-1α. As a result, we revealed an important mechanism for Parkin in keloid development and suggested that targeting Parkin could be an alternative method for keloid treatment.
Collapse
Affiliation(s)
- Rui Lei
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shizhen Zhang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Aiyu Liu
- Department of Neurology, Zhongda Hospital Affiliated to Southestern China University, Nanjing, China
| | - Xi Chen
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Wang
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Sun
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siya Dai
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghong Xu
- Department of Plastic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
63
|
Zilocchi M, Fasano M, Alberio T. Mitochondrial Proteins in the Development of Parkinson’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1158:17-44. [DOI: 10.1007/978-981-13-8367-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
64
|
Miyai T, Vasanth S, Melangath G, Deshpande N, Kumar V, Benischke AS, Chen Y, Price MO, Price FW, Jurkunas UV. Activation of PINK1-Parkin-Mediated Mitophagy Degrades Mitochondrial Quality Control Proteins in Fuchs Endothelial Corneal Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2061-2076. [PMID: 31361992 DOI: 10.1016/j.ajpath.2019.06.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 05/25/2019] [Accepted: 06/11/2019] [Indexed: 11/19/2022]
Abstract
Corneal endothelium (CE) is a monolayer of mitochondria-rich cells, critical for maintaining corneal transparency compatible with clear vision. Fuchs endothelial corneal dystrophy (FECD) is a heterogeneous, genetically complex disorder, where oxidative stress plays a key role in the rosette formation during the degenerative loss of CE. Increased mitochondrial fragmentation along with excessive mitophagy activation has been detected in FECD; however, the mechanism of aberrant mitochondrial dynamics in CE cell loss is poorly understood. Here, the role of oxidative stress in mitophagy activation in FECD is investigated. Immunoblotting of FECD ex vivo specimens revealed an accumulation of PINK1 and phospho-Parkin (Ser65) along with loss of total Parkin and total Drp1. Similarly, modeling of rosette formation with menadione (MN), led to phospho-Parkin accumulation in fragmented mitochondria resulting in mitophagy-induced mitochondrial clearance, albeit possibly in a PINK1-independent manner. Loss of PINK1, phospho-Drp1, and total Drp1 was prominent after MN-induced oxidative stress, but not after mitochondrial depolarization by carbonyl cyanide m-chlorophenyl hydrazone. Moreover, MN-induced mitophagy led to degradation of Parkin along with sequestration of Drp1 and PINK1 that was rescued by mitophagy inhibition. This study shows that in FECD, intracellular oxidative stress induces Parkin-mediated mitochondrial fragmentation where endogenous Drp1 and PINK1 are sequestered and degraded by mitophagy during degenerative loss of post-mitotic cells of ocular tissue.
Collapse
Affiliation(s)
- Takashi Miyai
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Shivakumar Vasanth
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Geetha Melangath
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Neha Deshpande
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Varun Kumar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Anne-Sophie Benischke
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Yuming Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | | | | | - Ula V Jurkunas
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
65
|
Mendonca P, Taka E, Soliman KFA. Proteomic analysis of the effect of the polyphenol pentagalloyl glucose on proteins involved in neurodegenerative diseases in activated BV‑2 microglial cells. Mol Med Rep 2019; 20:1736-1746. [PMID: 31257500 PMCID: PMC6625426 DOI: 10.3892/mmr.2019.10400] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 05/09/2019] [Indexed: 01/24/2023] Open
Abstract
Neuroinflammation and microglial activation are two important hallmarks of neurodegenerative diseases. Continuous microglial activation may cause the release of several cytotoxic molecules, including many cytokines that are involved in the inflammatory process. Therefore, attenuating inflammation caused by activated microglia may be an approach for the therapeutic management of neurodegenerative diseases. In addition, many studies have reported that polyphenol pentagalloyl glucose (1,2,3,4,6-penta-O-galloyl-β-D-glucose; PGG) is a molecule with potent anti-inflammatory effects, such as inhibiting the release of proinflammatory cytokines. Our previous studies revealed that PGG attenuated the expression of two inflammatory cytokines (murine monocyte chemoattractant protein-5 and pro-metalloproteinase-9) in lipopolysaccharide/interferon γ-activated BV-٢ microglial cells. Additionally, PGG modulated the NF-κB and MAPK signaling pathways by altering genes and proteins, which may affect the MAPK cascade and NF-κB activation. The aim of the present study was to investigate the ability of PGG to modulate the expression of proteins released in activated BV-2 microglial cells, which may be involved in the pathological process of inflammation and neurodegeneration. Proteomic analysis of activated BV-2 cells identified 17 proteins whose expression levels were significantly downregulated by PGG, including septin-7, ataxin-2, and adenylosuccinate synthetase isozyme 2 (ADSS). These proteins were previously described as being highly expressed in neurodegenerative diseases and/or involved in the signaling pathways associated with the formation and growth of neuronal connections and the control of Alzheimer's disease pathogenesis. The inhibitory effect of PGG on ataxin-2, septin-7 and ADSS was further confirmed at the protein and transcriptional levels. Therefore, the obtained results suggest that PGG, with its potent inhibitory effects on ataxin-2, septin-7 and ADSS, may have potential use in the therapeutic management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Patricia Mendonca
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Equar Taka
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Karam F A Soliman
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
66
|
Rinaldi L, Delle Donne R, Catalanotti B, Torres-Quesada O, Enzler F, Moraca F, Nisticò R, Chiuso F, Piccinin S, Bachmann V, Lindner HH, Garbi C, Scorziello A, Russo NA, Synofzik M, Stelzl U, Annunziato L, Stefan E, Feliciello A. Feedback inhibition of cAMP effector signaling by a chaperone-assisted ubiquitin system. Nat Commun 2019; 10:2572. [PMID: 31189917 PMCID: PMC6561907 DOI: 10.1038/s41467-019-10037-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Activation of G-protein coupled receptors elevates cAMP levels promoting dissociation of protein kinase A (PKA) holoenzymes and release of catalytic subunits (PKAc). This results in PKAc-mediated phosphorylation of compartmentalized substrates that control central aspects of cell physiology. The mechanism of PKAc activation and signaling have been largely characterized. However, the modes of PKAc inactivation by regulated proteolysis were unknown. Here, we identify a regulatory mechanism that precisely tunes PKAc stability and downstream signaling. Following agonist stimulation, the recruitment of the chaperone-bound E3 ligase CHIP promotes ubiquitylation and proteolysis of PKAc, thus attenuating cAMP signaling. Genetic inactivation of CHIP or pharmacological inhibition of HSP70 enhances PKAc signaling and sustains hippocampal long-term potentiation. Interestingly, primary fibroblasts from autosomal recessive spinocerebellar ataxia 16 (SCAR16) patients carrying germline inactivating mutations of CHIP show a dramatic dysregulation of PKA signaling. This suggests the existence of a negative feedback mechanism for restricting hormonally controlled PKA activities.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, 80131, Naples, Italy
| | - Rossella Delle Donne
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, 80131, Naples, Italy
| | - Bruno Catalanotti
- Department of Pharmacy, University Federico II, 80131, Naples, Italy
| | - Omar Torres-Quesada
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Florian Enzler
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Federica Moraca
- Department of Chemical Sciences, University Federico II, 80131, Naples, Italy
| | - Robert Nisticò
- European Brain Research Institute, Rita Levi-Montalcini Foundation and Department of Biology, University Tor Vergata, 00143, Rome, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, 80131, Naples, Italy
| | - Sonia Piccinin
- European Brain Research Institute, Rita Levi-Montalcini Foundation and Department of Biology, University Tor Vergata, 00143, Rome, Italy
| | - Verena Bachmann
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Herbert H Lindner
- Division of Clinical Biochemistry, Biocenter Medical University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Corrado Garbi
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, 80131, Naples, Italy
| | - Antonella Scorziello
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University Federico II, 80131, Naples, Italy
| | | | - Matthis Synofzik
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen and German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
| | - Ulrich Stelzl
- Institute of Pharmaceutical Sciences, University of Graz and BioTechMed-Graz, 8010, Graz, Austria
| | | | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University Federico II, 80131, Naples, Italy.
| |
Collapse
|
67
|
Park KR, Yun JS, Park MH, Jung YY, Yeo IJ, Nam KT, Kim HD, Song JK, Choi DY, Park PH, Han SB, Yun HM, Hong JT. Loss of parkin reduces lung tumor development by blocking p21 degradation. PLoS One 2019; 14:e0217037. [PMID: 31112565 PMCID: PMC6528990 DOI: 10.1371/journal.pone.0217037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 05/02/2019] [Indexed: 12/24/2022] Open
Abstract
Several epidemiological studies have demonstrated the reciprocal relationship between the development of cancer and Parkinson’s disease (PD). However, the possible mechanisms underlying this relationship remain unclear. To identify this relationship, we first compared lung tumor growth in parkin knockout (KO) mice and wild-type (WT) mice. Parkin KO mice showed decreased lung tumor growth and increased expression of p21, a cell cycle arrester, as compared with WT mice. We also found that parkin interacts with p21, resulting in its degradation; however, parkin KO, knockdown, as well as mutation (R275W or G430D) reduced the degradation of p21. We investigated whether parkin KO increases the association of p21 with proliferating cell nuclear antigen (PCNA) or CDK2 by reducing p21 degradation, and, thus, arresting the cell cycle. The interaction between p21 and PCNA or CDK2 was also enhanced by parkin knockdown, and this increased interaction induced sub G0/G1 arrest, leading to cell death. Therefore, our data indicate that parkin KO reduces the development of lung tumors via cell cycle arrest by blocking the degradation of p21. These findings suggest that PD could be associated with lower lung cancer incidence.
Collapse
Affiliation(s)
- Kyung-Ran Park
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Dongdaemun-Gu, Seoul, Republic of Korea
| | - Jae Suk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Yu Yeon Jung
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Kyung Tak Nam
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Hae Deun Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Ju Kyoung Song
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Dongdaemun-Gu, Seoul, Republic of Korea
- * E-mail: (JTH); (HMY)
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
- * E-mail: (JTH); (HMY)
| |
Collapse
|
68
|
Liu X, Hebron M, Shi W, Lonskaya I, Moussa CEH. Ubiquitin specific protease-13 independently regulates parkin ubiquitination and alpha-synuclein clearance in alpha-synucleinopathies. Hum Mol Genet 2019; 28:548-560. [PMID: 30329047 DOI: 10.1093/hmg/ddy365] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022] Open
Abstract
Ubiquitin specific proteases (USPs) are de-ubiquitinases (DUBs) that control protein ubiquitination cycle. The role of DUBs is poorly understood in neurodegenerative diseases. We found that USP13 is overexpressed in post-mortem Parkinson's disease (PD) brains. We investigated whether changes in USP13 levels can affect two molecules, parkin and alpha-synuclein, that are implicated in PD pathogenesis. Parkin is an E3 ubiquitin ligase that is regulated by ubiquitination and targets certain proteins for degradation, and alpha-synuclein may be ubiquitinated and recycled in the normal brain. We found that USP13 independently regulates parkin and alpha-synuclein ubiquitination in models of alpha-synucleinopathies. USP13 shRNA knockdown increases alpha-synuclein ubiquitination and clearance, in a parkin-independent manner. Furthermore, USP13 overexpression counteracts the effects of a tyrosine kinase inhibitor, Nilotinib, while USP13 knockdown facilitates Nilotinib effects on alpha-synculein clearance, suggesting that alpha-synuclein ubiquitnation is important for its clearance. These studies provide novel evidence of USP13 effects on parkin and alpha-synuclein metabolism and suggest that USP13 is a potential therapeutic target in the alpha-synucleinopathies.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Michaeline Hebron
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Wangke Shi
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Irina Lonskaya
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| |
Collapse
|
69
|
Dombi E, Mortiboys H, Poulton J. Modulating Mitophagy in Mitochondrial Disease. Curr Med Chem 2019; 25:5597-5612. [DOI: 10.2174/0929867324666170616101741] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 01/28/2023]
Abstract
Mitochondrial diseases may result from mutations in the maternally-inherited mitochondrial
DNA (mtDNA) or from mutations in nuclear genes encoding mitochondrial proteins.
Their bi-genomic nature makes mitochondrial diseases a very heterogeneous group of
disorders that can present at any age and can affect any type of tissue.
The autophagic-lysosomal degradation pathway plays an important role in clearing dysfunctional
and redundant mitochondria through a specific quality control mechanism termed mitophagy.
Mitochondria could be targeted for autophagic degradation for a variety of reasons including
basal turnover for recycling, starvation induced degradation, and degradation due to
damage. While the core autophagic machinery is highly conserved and common to most
pathways, the signaling pathways leading to the selective degradation of damaged mitochondria
are still not completely understood. Type 1 mitophagy due to nutrient starvation is dependent
on PI3K (phosphoinositide 3-kinase) for autophagosome formation but independent
of mitophagy proteins, PINK1 (PTEN-induced putative kinase 1) and Parkin. Whereas type 2
mitophagy that occurs due to damage is dependent on PINK1 and Parkin but does not require
PI3K.
Autophagy and mitophagy play an important role in human disease and hence could serve as
therapeutic targets for the treatment of mitochondrial as well as neurodegenerative disorders.
Therefore, we reviewed drugs that are known modulators of autophagy (AICAR and metformin)
and may affect this by activating the AMP-activated protein kinase signaling pathways.
Furthermore, we reviewed the data available on supplements, such as Coenzyme Q and
the quinone idebenone, that we assert rescue increased mitophagy in mitochondrial disease by
benefiting mitochondrial function.
Collapse
Affiliation(s)
- Eszter Dombi
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, Neuroscience Department, University of Sheffield, United Kingdom
| | - Joanna Poulton
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
70
|
Kappel S, Borgström A, Stokłosa P, Dörr K, Peinelt C. Store-operated calcium entry in disease: Beyond STIM/Orai expression levels. Semin Cell Dev Biol 2019; 94:66-73. [PMID: 30630032 DOI: 10.1016/j.semcdb.2019.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/29/2018] [Accepted: 01/05/2019] [Indexed: 12/19/2022]
Abstract
Precise intracellular calcium signaling is crucial to numerous cellular functions. In non-excitable cells, store-operated calcium entry (SOCE) is a key step in the generation of intracellular calcium signals. Tight regulation of SOCE is important, and dysregulation is involved in several pathophysiological cellular malfunctions. The current underlying SOCE, calcium release-activated calcium current (ICRAC), was first discovered almost three decades ago. Since its discovery, the molecular components of ICRAC, Orai1 and stromal interaction molecule 1 (STIM1), have been extensively investigated. Several regulatory mechanisms and proteins contribute to alterations in SOCE and cellular malfunctions in cancer, immune and neurodegenerative diseases, inflammation, and neuronal disorders. This review summarizes these regulatory mechanisms, including glycosylation, pH sensing, and the regulatory proteins golli, α-SNAP, SARAF, ORMDL3, CRACR2A, and TRPM4 channels.
Collapse
Affiliation(s)
- Sven Kappel
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Anna Borgström
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Paulina Stokłosa
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | | | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| |
Collapse
|
71
|
Tan SH, Karri V, Tay NWR, Chang KH, Ah HY, Ng PQ, Ho HS, Keh HW, Candasamy M. Emerging pathways to neurodegeneration: Dissecting the critical molecular mechanisms in Alzheimer's disease, Parkinson's disease. Biomed Pharmacother 2019; 111:765-777. [PMID: 30612001 DOI: 10.1016/j.biopha.2018.12.101] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 01/02/2023] Open
Abstract
Neurodegenerative diseases are usually sporadic in nature and commonly influenced by a wide range of genetic, life style and environmental factors. A unifying feature of Alzheimer's disease (AD) and Parkinson's disease (PD) is the abnormal accumulation and processing of mutant or damaged intra and extracellular proteins; this leads to neuronal vulnerability and dysfunction in the brain. Through a detailed review of ubiquitin proteasome, mRNA splicing, mitochondrial dysfunction, and oxidative stress pathway interrelation on neurodegeneration can improve the understanding of the disease mechanism. The identified pathways common to AD and PD nominate promising new targets for further studies, and as well as biomarkers. These insights suggested would likely provide major stimuli for developing unified treatment approaches to combat neurodegeneration. More broadly, pathways can serve as vehicles for integrating findings from diverse studies of neurodegeneration. The evidence examined in this review provides a brief overview of the current literature on significant pathways in promoting in AD, PD. Additionally, these insights suggest that biomarkers and treatment strategies may require simultaneous targeting of multiple components.
Collapse
Affiliation(s)
- Sean Hong Tan
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Venkatanaidu Karri
- Department of Toxicogenomics, Faculty of Health, Medicines, Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Nicole Wuen Rong Tay
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Kuan Hui Chang
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hui Yen Ah
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Phui Qi Ng
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hui San Ho
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hsiao Wai Keh
- School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
72
|
Liu X, Hebron ML, Mulki S, Wang C, Lekah E, Ferrante D, Shi W, Kurd-Misto B, Moussa C. Ubiquitin Specific Protease 13 Regulates Tau Accumulation and Clearance in Models of Alzheimer's Disease. J Alzheimers Dis 2019; 72:425-441. [PMID: 31594232 DOI: 10.3233/jad-190635] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ubiquitin Specific Protease-13 (USP13) is a de-ubiquinating enzyme that regulates protein ubiquitination and clearance. The role of USP13 is largely unknown in neurodegeneration. In this study we aim to demonstrate whether tau accumulation and/or clearance depends on ubiquitination/de-ubiquitination via USP-13. We used transgenic animal models of human amyloid precursor protein (APP) or P301L tau mutations and genetically knocked-down USP13 expression via shRNA to determine USP13 effects on tau ubiquitination and levels. We found a two-fold increase of USP13 levels in postmortem Alzheimer's disease (AD) brains. USP13 knockdown significantly increased the activity of the 20S proteasome and reduced the levels of hyper-phosphorylated tau (p-tau) in primary cortical neurons. USP13 knockdown also reduced the levels of amyloid and increased p-tau ubiquitination and clearance in transgenic animal models that overexpress murine tau as a result of the expression of familial APP mutations (TgAPP) and the human mutant P301L tau (rTg4510), respectively. Clearance of p-tau appears to be mediated by autophagy in these animal models. Taken together, these data suggest that USP13 knockdown reduces p-tau accumulation via regulation of ubiquitination/de-ubiquitination and mediates its clearance via autophagy and/or the proteasome. These results suggest that USP13 inhibition may be a therapeutic strategy to reduce accumulation of plaques and toxic p-tau in AD and human tauopathies.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Sanjana Mulki
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Chen Wang
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Elizabeth Lekah
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Dalila Ferrante
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Wangke Shi
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Bahjat Kurd-Misto
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
73
|
Lin CH, Shen YR, Wang HY, Chiang CW, Wang CY, Kuo PL. Regulation of septin phosphorylation: SEPT12 phosphorylation in sperm septin assembly. Cytoskeleton (Hoboken) 2018; 76:137-142. [PMID: 30160375 DOI: 10.1002/cm.21491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 08/08/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022]
Abstract
The sperm annulus, a septin-based ring structure, is important for reproductive physiology. It is composed of SEPT12-based septin core complex followed by assembling as octameric filament. In clinical examinations, mutations of Septin12 result in male infertility, immotile sperm, as well as sperm with defective annuli. The dynamic assembly of septin filaments is regulated by several post-translational modifications, including sumoylation, acetylation, and phosphorylation. Here, we briefly review the biological significance and the regulation of SEPT12 phosphorylation in the mammalian sperm physiology. During mammalian spermiogenesis, the phosphorylation of SEPT12 on Ser198 residue is important in regulating mammalian annulus architectures. SEPT12 phosphomimetic knock-in mice displayed poor male fertility due to weak sperm motility and loss of the sperm annulus. SEPT12 is phosphorylated via Protein kinase A (PKA), and its phosphorylation interfered with SEPT12 polymerization into complexes and filaments. Taken together, the phosphorylation status of SEPT12 is crucial for its function in regulating the mammalian sperm physiology.
Collapse
Affiliation(s)
- Chun-Hsiang Lin
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ru Shen
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Han-Yu Wang
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Obstetrics and Gynecology, National Cheng-Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
74
|
Akhmetova KA, Chesnokov IN, Fedorova SA. [Functional Characterization of Septin Complexes]. Mol Biol (Mosk) 2018; 52:155-171. [PMID: 29695686 DOI: 10.7868/s0026898418020015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/27/2016] [Indexed: 12/28/2022]
Abstract
Septins belong to a family of conserved GTP-binding proteins found in majority of eukaryotic species except for higher plants. Septins form nonpolar complexes that further polymerize into filaments and associate with cell membranes, thus comprising newly acknowledged cytoskeletal system. Septins participate in a variety of cell processes and contribute to various pathophysiological states, including tumorigenesis and neurodegeneration. Here, we review the structural and functional properties of septins and the regulation of their dynamics with special emphasis on the role of septin filaments as a cytoskeletal system and its interaction with actin and microtubule cytoskeletons. We also discuss how septins compartmentalize the cell by forming local protein-anchoring scaffolds and by providing barriers for the lateral diffusion of the membrane proteins.
Collapse
Affiliation(s)
- K A Akhmetova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia.,University of Alabama at Birmingham, Birmingham, 35294 USA.,Novosibirsk National Research State University, Novosibirsk, 630090 Russia
| | - I N Chesnokov
- University of Alabama at Birmingham, Birmingham, 35294 USA
| | - S A Fedorova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia.,Novosibirsk National Research State University, Novosibirsk, 630090 Russia.,
| |
Collapse
|
75
|
Presynaptic Dysfunction by Familial Factors in Parkinson Disease. Int Neurourol J 2018; 22:S115-121. [PMID: 30396260 PMCID: PMC6234725 DOI: 10.5213/inj.1836216.108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/10/2018] [Indexed: 12/28/2022] Open
Abstract
Parkinson disease (PD) is the second most prevalent neurodegenerative disorder after Alzheimer disease. The loss of specific brain area, the substantia nigra pars compacta is known as a major etiology, however it is not fully understood how this neurodegeneration is initiated and what precisely causes this disease. As one aspect of pathophysiology for PD, synaptic dysfunction (synaptopathy) is thought to be an earlier appearance for neurodegeneration. In addition, some of the familial factors cumulatively exhibit that these factors such as α-synuclein, leucine-rich repeat kinase 2, parkin, PTEN-induced kinase 1, and DJ-1 are involved in the regulation of synaptic function and missense mutants of familial factors found in PD-patient show dysregulation of synaptic functions. In this review, we have discussed the physiological function of these genetic factors in presynaptic terminal and how dysregulation of presynaptic function by genetic factors might be related to the pathogenesis of Parkinson disease.
Collapse
|
76
|
Lee YS, Jung YY, Park MH, Yeo IJ, Im HS, Nam KT, Kim HD, Kang SK, Song JK, Kim YR, Choi DY, Park PH, Han SB, Yun JS, Hong JT. Deficiency of parkin suppresses melanoma tumor development and metastasis through inhibition of MFN2 ubiquitination. Cancer Lett 2018; 433:156-164. [DOI: 10.1016/j.canlet.2018.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 06/30/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022]
|
77
|
Zhu M, Cortese GP, Waites CL. Parkinson's disease-linked Parkin mutations impair glutamatergic signaling in hippocampal neurons. BMC Biol 2018; 16:100. [PMID: 30200940 PMCID: PMC6130078 DOI: 10.1186/s12915-018-0567-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 08/24/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD)-associated E3 ubiquitin ligase Parkin is enriched at glutamatergic synapses, where it ubiquitinates multiple substrates, suggesting that its mutation/loss-of-function could contribute to the etiology of PD by disrupting excitatory neurotransmission. Here, we evaluate the impact of four common PD-associated Parkin point mutations (T240M, R275W, R334C, G430D) on glutamatergic synaptic function in hippocampal neurons. RESULTS We find that expression of these point mutants in cultured hippocampal neurons from Parkin-deficient and Parkin-null backgrounds alters NMDA and AMPA receptor-mediated currents and cell-surface levels and prevents the induction of long-term depression. Mechanistically, we demonstrate that Parkin regulates NMDA receptor trafficking through its ubiquitination of GluN1, and that all four mutants are impaired in this ubiquitinating activity. Furthermore, Parkin regulates synaptic AMPA receptor trafficking via its binding and retention of the postsynaptic scaffold Homer1, and all mutants are similarly impaired in this capacity. CONCLUSION Our findings demonstrate that pathogenic Parkin mutations disrupt glutamatergic synaptic transmission in hippocampal neurons by impeding NMDA and AMPA receptor trafficking. Such effects may contribute to the pathophysiology of PD in PARK2 patients.
Collapse
Affiliation(s)
- Mei Zhu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032 USA
| | - Giuseppe P. Cortese
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032 USA
- Department of Psychiatry, Columbia University Medical Center, New York, NY USA
| | - Clarissa L. Waites
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032 USA
- Department of Neuroscience, Columbia University, New York, NY USA
- Waites Lab, 650 W. 168th St. Black Building 1210B, New York, NY 10032 USA
| |
Collapse
|
78
|
Lizama BN, Palubinsky AM, McLaughlin B. Alterations in the E3 ligases Parkin and CHIP result in unique metabolic signaling defects and mitochondrial quality control issues. Neurochem Int 2018; 117:139-155. [PMID: 28851515 PMCID: PMC5826822 DOI: 10.1016/j.neuint.2017.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 08/11/2017] [Accepted: 08/21/2017] [Indexed: 01/07/2023]
Abstract
E3 ligases are essential scaffold proteins, facilitating the transfer of ubiquitin from E2 enzymes to lysine residues of client proteins via isopeptide bonds. The specificity of substrate binding and the expression and localization of E3 ligases can, however, endow these proteins with unique features with variable effects on mitochondrial, metabolic and CNS function. By comparing and contrasting two E3 ligases, Parkin and C-terminus of HSC70-Interacting protein (CHIP) we seek to highlight the biophysical properties that may promote mitochondrial dysfunction, acute stress signaling and critical developmental periods to cease in response to mutations in these genes. Encoded by over 600 human genes, RING-finger proteins are the largest class of E3 ligases. Parkin contains three RING finger domains, with R1 and R2 separated by an in-between region (IBR) domain. Loss-of-function mutations in Parkin were identified in patients with early onset Parkinson's disease. CHIP is a member of the Ubox family of E3 ligases. It contains an N-terminal TPR domain and forms unique asymmetric homodimers. While CHIP can substitute for mutated Parkin and enhance survival, CHIP also has unique functions. The differences between these proteins are underscored by the observation that unlike Parkin-deficient animals, CHIP-null animals age prematurely and have significantly impaired motor function. These properties make these E3 ligases appealing targets for clinical intervention. In this work, we discuss how biophysical and metabolic properties of these E3 ligases have driven rapid progress in identifying roles for E3 ligases in development, proteostasis, mitochondrial biology, and cell health, as well as new data about how these proteins alter the CNS proteome.
Collapse
Affiliation(s)
- Britney N Lizama
- Neuroscience Graduate Group, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Vanderbilt Brain Institute, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States.
| | - Amy M Palubinsky
- Neuroscience Graduate Group, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Vanderbilt Brain Institute, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States
| | - BethAnn McLaughlin
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Department of Neurology, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States; Department of Pharmacology, Vanderbilt University Medical Center, 465 21st Ave S MRB III, Nashville, TN 37240, United States
| |
Collapse
|
79
|
Liu J, Zhang C, Hu W, Feng Z. Parkinson's disease-associated protein Parkin: an unusual player in cancer. Cancer Commun (Lond) 2018; 38:40. [PMID: 29941042 PMCID: PMC6020249 DOI: 10.1186/s40880-018-0314-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
The mutation of the Parkin gene is a cause of familial Parkinson’s disease. A growing body of evidence suggests that Parkin also functions as a tumor suppressor. Parkin is an ubiquitin E3 ligase, and plays important roles in a variety of cellular processes implicated in tumorigenesis, including cell cycle, cell proliferation, apoptosis, metastasis, mitophagy and metabolic reprogramming. Here we review the role and mechanism of Parkin in cancer.
Collapse
Affiliation(s)
- Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Cen Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, 08903, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, 08903, USA. .,Department of Pharmacology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, 08903, USA.
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, 08903, USA. .,Department of Pharmacology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
80
|
Hiroi N. Critical reappraisal of mechanistic links of copy number variants to dimensional constructs of neuropsychiatric disorders in mouse models. Psychiatry Clin Neurosci 2018; 72:301-321. [PMID: 29369447 PMCID: PMC5935536 DOI: 10.1111/pcn.12641] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/27/2017] [Accepted: 01/19/2018] [Indexed: 12/17/2022]
Abstract
Copy number variants are deletions and duplications of a few thousand to million base pairs and are associated with extraordinarily high levels of autism spectrum disorder, schizophrenia, intellectual disability, or attention-deficit hyperactivity disorder. The unprecedented levels of robust and reproducible penetrance of copy number variants make them one of the most promising and reliable entry points to delve into the mechanistic bases of many mental disorders. However, the precise mechanistic bases of these associations still remain elusive in humans due to the many genes encoded in each copy number variant and the diverse associated phenotypic features. Genetically engineered mice have provided a technical means to ascertain precise genetic mechanisms of association between copy number variants and dimensional aspects of mental illnesses. Molecular, cellular, and neuronal phenotypes can be detected as potential mechanistic substrates for various behavioral constructs of mental illnesses. However, mouse models come with many technical pitfalls. Genetic background is not well controlled in many mouse models, leading to rather obvious interpretative issues. Dose alterations of many copy number variants and single genes within copy number variants result in some molecular, cellular, and neuronal phenotypes without a behavioral phenotype or with a behavioral phenotype opposite to what is seen in humans. In this review, I discuss technical and interpretative pitfalls of mouse models of copy number variants and highlight well-controlled studies to suggest potential neuronal mechanisms of dimensional aspects of mental illnesses. Mouse models of copy number variants represent toeholds to achieve a better understanding of the mechanistic bases of dimensions of neuropsychiatric disorders and thus for development of mechanism-based therapeutic options in humans.
Collapse
Affiliation(s)
- Noboru Hiroi
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, USA.,Department of Neuroscience, Albert Einstein College of Medicine, New York, USA.,Department of Genetics, Albert Einstein College of Medicine, New York, USA
| |
Collapse
|
81
|
Sasidharakurup H, Melethadathil N, Nair B, Diwakar S. A Systems Model of Parkinson's Disease Using Biochemical Systems Theory. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:454-464. [PMID: 28816645 DOI: 10.1089/omi.2017.0056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder, affects millions of people and has gained attention because of its clinical roles affecting behaviors related to motor and nonmotor symptoms. Although studies on PD from various aspects are becoming popular, few rely on predictive systems modeling approaches. Using Biochemical Systems Theory (BST), this article attempts to model and characterize dopaminergic cell death and understand pathophysiology of progression of PD. PD pathways were modeled using stochastic differential equations incorporating law of mass action, and initial concentrations for the modeled proteins were obtained from literature. Simulations suggest that dopamine levels were reduced significantly due to an increase in dopaminergic quinones and 3,4-dihydroxyphenylacetaldehyde (DOPAL) relating to imbalances compared to control during PD progression. Associating to clinically observed PD-related cell death, simulations show abnormal parkin and reactive oxygen species levels with an increase in neurofibrillary tangles. While relating molecular mechanistic roles, the BST modeling helps predicting dopaminergic cell death processes involved in the progression of PD and provides a predictive understanding of neuronal dysfunction for translational neuroscience.
Collapse
Affiliation(s)
| | - Nidheesh Melethadathil
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham (Amrita University) , Kollam, India
| | - Bipin Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham (Amrita University) , Kollam, India
| | - Shyam Diwakar
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham (Amrita University) , Kollam, India
| |
Collapse
|
82
|
|
83
|
Wilkaniec A, Lenkiewicz AM, Czapski GA, Jęśko HM, Hilgier W, Brodzik R, Gąssowska-Dobrowolska M, Culmsee C, Adamczyk A. Extracellular Alpha-Synuclein Oligomers Induce Parkin S-Nitrosylation: Relevance to Sporadic Parkinson's Disease Etiopathology. Mol Neurobiol 2018; 56:125-140. [PMID: 29681024 PMCID: PMC6334739 DOI: 10.1007/s12035-018-1082-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/11/2018] [Indexed: 01/10/2023]
Abstract
α-Synuclein (ASN) and parkin, a multifunctional E3 ubiquitin ligase, are two proteins that are associated with the pathophysiology of Parkinson’s disease (PD). Excessive release of ASN, its oligomerization, aggregation, and deposition in the cytoplasm contribute to neuronal injury and cell death through oxidative-nitrosative stress induction, mitochondrial impairment, and synaptic dysfunction. In contrast, overexpression of parkin provides protection against cellular stresses and prevents dopaminergic neural cell loss in several animal models of PD. However, the influence of ASN on the function of parkin is largely unknown. Therefore, the aim of this study was to investigate the effect of extracellular ASN oligomers on parkin expression, S-nitrosylation, as well as its activity. For these investigations, we used rat pheochromocytoma (PC12) cell line treated with exogenous oligomeric ASN as well as PC12 cells with parkin overexpression and parkin knock-down. The experiments were performed using spectrophotometric, spectrofluorometric, and immunochemical methods. We found that exogenous ASN oligomers induce oxidative/nitrosative stress leading to parkin S-nitrosylation. Moreover, this posttranslational modification induced the elevation of parkin autoubiquitination and degradation of the protein. The decreased parkin levels resulted in significant cell death, whereas parkin overexpression protected against toxicity induced by extracellular ASN oligomers. We conclude that lowering parkin levels by extracellular ASN may significantly contribute to the propagation of neurodegeneration in PD pathology through accumulation of defective proteins as a consequence of parkin degradation.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland
| | - Anna M Lenkiewicz
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland
| | - Henryk M Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland
| | - Wojciech Hilgier
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland
| | | | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043, Marburg, Germany
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5 Street, 02-106, Warsaw, Poland.
| |
Collapse
|
84
|
Sheehan P, Yue Z. Deregulation of autophagy and vesicle trafficking in Parkinson's disease. Neurosci Lett 2018; 697:59-65. [PMID: 29627340 DOI: 10.1016/j.neulet.2018.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 04/03/2018] [Accepted: 04/04/2018] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized pathologically by the selective loss of dopaminergic neurons in the substantia nigra and the intracellular accumulation of α-synuclein in the Lewy bodies. While the pathogenic mechanisms of PD are poorly understood, many lines of evidence point to a role of altered autophagy and membrane trafficking in the development of the disease. Emerging studies show that connections between the deregulation of autophagy and synaptic vesicle (SV) trafficking may contribute to PD. Here we review the evidence that many PD related-genes have roles in both autophagy and SV trafficking and examine how deregulation of these pathways contributes to PD pathogenesis. This review also discusses recent studies aimed at uncovering the role of PD-linked genes in autophagy-lysosome function.
Collapse
Affiliation(s)
- Patricia Sheehan
- Department of Neurology, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Zhenyu Yue
- Department of Neurology, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, USA.
| |
Collapse
|
85
|
Abstract
Synapse is the basic structural and functional component for neural communication in the brain. The presynaptic terminal is the structural and functionally essential area that initiates communication and maintains the continuous functional neural information flow. It contains synaptic vesicles (SV) filled with neurotransmitters, an active zone for release, and numerous proteins for SV fusion and retrieval. The structural and functional synaptic plasticity is a representative characteristic; however, it is highly vulnerable to various pathological conditions. In fact, synaptic alteration is thought to be central to neural disease processes. In particular, the alteration of the structural and functional phenotype of the presynaptic terminal is a highly significant evidence for neural diseases. In this review, we specifically describe structural and functional alteration of nerve terminals in several neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD).
Collapse
Affiliation(s)
- Jae Ryul Bae
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Sung Hyun Kim
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
86
|
Williams JA, Ding WX. Mechanisms, pathophysiological roles and methods for analyzing mitophagy - recent insights. Biol Chem 2018; 399:147-178. [PMID: 28976892 DOI: 10.1515/hsz-2017-0228] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
In 2012, we briefly summarized the mechanisms, pathophysiological roles and methods for analyzing mitophagy. As then, the mitophagy field has continued to grow rapidly, and many new molecular mechanisms regulating mitophagy and molecular tools for monitoring mitophagy have been discovered and developed. Therefore, the purpose of this review is to update information regarding these advances in mitophagy while focusing on basic molecular mechanisms of mitophagy in different organisms and its pathophysiological roles. We also discuss the advantage and limitations of current methods to monitor and quantify mitophagy in cultured cells and in vivo mouse tissues.
Collapse
Affiliation(s)
- Jessica A Williams
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
87
|
Zhong P, Hu Z, Jiang H, Yan Z, Feng J. Dopamine Induces Oscillatory Activities in Human Midbrain Neurons with Parkin Mutations. Cell Rep 2018; 19:1033-1044. [PMID: 28467897 PMCID: PMC5492970 DOI: 10.1016/j.celrep.2017.04.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 01/23/2017] [Accepted: 04/07/2017] [Indexed: 12/18/2022] Open
Abstract
Locomotor symptoms in Parkinson's disease (PD) are accompanied by widespread oscillatory neuronal activities in basal ganglia. Here, we show that activation of dopamine D1-class receptors elicits a large rhythmic bursting of spontaneous excitatory postsynaptic currents (sEPSCs) in midbrain neurons differentiated from induced pluripotent stem cells (iPSCs) of PD patients with parkin mutations, but not normal subjects. Overexpression of wild-type parkin, but not its PD-causing mutant, abolishes the oscillatory activities in patient neurons. Dopamine induces a delayed enhancement in the amplitude of spontaneous, but not miniature, EPSCs, thus increasing quantal content. The results suggest that presynaptic regulation of glutamatergic transmission by dopamine D1-class receptors is significantly potentiated by parkin mutations. The aberrant dopaminergic regulation of presynaptic glutamatergic transmission in patient-specific iPSC-derived midbrain neurons provides a mechanistic clue to PD pathophysiology, and it demonstrates the usefulness of this model system in understanding how mutations of parkin cause movement symptoms in Parkinson's disease.
Collapse
Affiliation(s)
- Ping Zhong
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA; Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Zhixing Hu
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA; Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Houbo Jiang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA; Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA; Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA.
| | - Jian Feng
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA; Veterans Affairs Western New York Healthcare System, Buffalo, NY 14215, USA.
| |
Collapse
|
88
|
Do YJ, Yun SY, Park MY, Kim E. The M458L missense mutation disrupts the catalytic properties of Parkin. FEBS Lett 2017; 592:78-88. [PMID: 29223129 DOI: 10.1002/1873-3468.12934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/21/2017] [Accepted: 11/30/2017] [Indexed: 11/09/2022]
Abstract
Parkin encodes an E3 ubiquitin ligase, and mutations affecting its catalytic potential are implicated in autosomal recessive Parkinson's disease (PD). The M458L mutation of parkin and its enzymatic effects require characterization. Therefore, we examined the enzymatic activity of Parkin with M458L mutation. We show that the M458L mutant retains its autoubiquitination potential in vitro but not in cells. Fas-associated factor 1 and p38 (substrates of Parkin) are able to bind to the M458L mutant in cells; however, these Parkin substrates are not ubiquitinated and degraded in M458L mutant-transfected cells. Moreover, M458L mutant fails to protect the mitochondria against hydrogen peroxide, leading to cell death. Considering the role of mitochondrial dysfunction in PD pathogenesis, our results imply a causative role for the M458L mutation in neurodegeneration.
Collapse
Affiliation(s)
- Yun-Ju Do
- Department of Biological Sciences, Chungnam National University, Daejeon, Korea
| | - Seo Young Yun
- Department of Biological Sciences, Chungnam National University, Daejeon, Korea
| | - Min-Young Park
- Department of Biological Sciences, Chungnam National University, Daejeon, Korea
| | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, Korea
| |
Collapse
|
89
|
Parkin targets HIF-1α for ubiquitination and degradation to inhibit breast tumor progression. Nat Commun 2017; 8:1823. [PMID: 29180628 PMCID: PMC5703960 DOI: 10.1038/s41467-017-01947-w] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 10/27/2017] [Indexed: 02/08/2023] Open
Abstract
Mutations in E3 ubiquitin ligase Parkin have been linked to familial Parkinson’s disease. Accumulating evidence suggests that Parkin is a tumor suppressor, but the underlying mechanism is poorly understood. Here we show that Parkin is an E3 ubiquitin ligase for hypoxia-inducible factor 1α (HIF-1α). Parkin interacts with HIF-1α and promotes HIF-1α degradation through ubiquitination, which in turn inhibits metastasis of breast cancer cells. Parkin downregulation in breast cancer cells promotes metastasis, which can be inhibited by targeting HIF-1α with RNA interference or the small-molecule inhibitor YC-1. We further identify lysine 477 (K477) of HIF-1α as a major ubiquitination site for Parkin. K477R HIF-1α mutation and specific cancer-associated Parkin mutations largely abolish the functions of Parkin to ubiquitinate HIF-1α and inhibit cancer metastasis. Importantly, Parkin expression is inversely correlated with HIF-1α expression and metastasis in breast cancer. Our results reveal an important mechanism for Parkin in tumor suppression and HIF-1α regulation. Parkin is an E3 ubiquitin ligase involved in Parkinson’s disease. Parkin has also been linked to cancer suppression but the mechanisms are unclear. Here the authors show that Parkin regulates HIF-1α through ubiquitin-dependent degradation, thus inhibiting metastasis of breast cancer cells.
Collapse
|
90
|
Sassone J, Serratto G, Valtorta F, Silani V, Passafaro M, Ciammola A. The synaptic function of parkin. Brain 2017; 140:2265-2272. [PMID: 28335015 DOI: 10.1093/brain/awx006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022] Open
Abstract
Loss of function mutations in the gene PARK2, which encodes the protein parkin, cause autosomal recessive juvenile parkinsonism, a neurodegenerative disease characterized by degeneration of the dopaminergic neurons localized in the substantia nigra pars compacta. No therapy is effective in slowing disease progression mostly because the pathogenesis of the disease is yet to be understood. From accruing evidence suggesting that the protein parkin directly regulates synapses it can be hypothesized that PARK2 gene mutations lead to early synaptic damage that results in dopaminergic neuron loss over time. We review evidence that supports the role of parkin in modulating excitatory and dopaminergic synapse functions. We also discuss how these findings underpin the concept that autosomal recessive juvenile parkinsonism can be primarily a synaptopathy. Investigation into the molecular interactions between parkin and synaptic proteins may yield novel targets for pharmacologic interventions.
Collapse
Affiliation(s)
- Jenny Sassone
- San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - GiuliaMaia Serratto
- CNR Institute of Neuroscience, Department BIOMETRA, Università degli Studi di Milano, Milan, Italy.,IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, Milan, Italy
| | - Flavia Valtorta
- San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy
| | - Vincenzo Silani
- IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, Milan, Italy.,Department of Pathophysiology and Transplantation, 'Dino Ferrari' Centre, Università degli Studi di Milano, Milan, Italy
| | - Maria Passafaro
- CNR Institute of Neuroscience, Department BIOMETRA, Università degli Studi di Milano, Milan, Italy
| | - Andrea Ciammola
- IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, Milan, Italy
| |
Collapse
|
91
|
Activation mechanisms of the E3 ubiquitin ligase parkin. Biochem J 2017; 474:3075-3086. [PMID: 28860335 DOI: 10.1042/bcj20170476] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
Abstract
Monogenetic, familial forms of Parkinson's disease (PD) only account for 5-10% of the total number of PD cases, but analysis of the genes involved therein is invaluable to understanding PD-associated neurodegenerative signaling. One such gene, parkin, encodes a 465 amino acid E3 ubiquitin ligase. Of late, there has been considerable interest in the role of parkin signaling in PD and in identifying its putative substrates, as well as the elucidation of the mechanisms through which parkin itself is activated. Its dysfunction underlies both inherited and idiopathic PD-associated neurodegeneration. Here, we review recent literature that provides a model of activation of parkin in the setting of mitochondrial damage that involves PINK1 (PTEN-induced kinase-1) and phosphoubiquitin. We note that neuronal parkin is primarily a cytosolic protein (with various non-mitochondrial functions), and discuss potential cytosolic parkin activation mechanisms.
Collapse
|
92
|
Dove KK, Klevit RE. RING-Between-RING E3 Ligases: Emerging Themes amid the Variations. J Mol Biol 2017; 429:3363-3375. [PMID: 28827147 DOI: 10.1016/j.jmb.2017.08.008] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 11/30/2022]
Abstract
Covalent, reversible, post-translational modification of cellular proteins with the small modifier, ubiquitin (Ub), regulates virtually every known cellular process in eukaryotes. The process is carried out by a trio of enzymes: a Ub-activating (E1) enzyme, a Ub-conjugating (E2) enzyme, and a Ub ligase (E3) enzyme. RING-in-Between-RING (RBR) E3s constitute one of three classes of E3 ligases and are defined by a RING-HECT-hybrid mechanism that utilizes a E2-binding RING domain and a second domain (called RING2) that contains an active site Cys required for the formation of an obligatory E3~Ub intermediate. Albeit a small class, RBR E3s in humans regulate diverse cellular process. This review focuses on non-Parkin members such as HOIP/HOIL-1L (the only E3s known to generate linear Ub chains), HHARI and TRIAD1, both of which have been recently demonstrated to work together with Cullin RING E3 ligases. We provide a brief historical background and highlight, summarize, and discuss recent developments in the young field of RBR E3s. Insights reviewed here include new understandings of the RBR Ub-transfer mechanism, specifically the role of RING1 and various Ub-binding sites, brief structural comparisons among members, and different modes of auto-inhibition and activation.
Collapse
Affiliation(s)
- Katja K Dove
- Department of Biochemistry, University of Washington, Seattle, WA, United States
| | - Rachel E Klevit
- Department of Biochemistry, University of Washington, Seattle, WA, United States.
| |
Collapse
|
93
|
Parkin Deficiency Reduces Hippocampal Glutamatergic Neurotransmission by Impairing AMPA Receptor Endocytosis. J Neurosci 2017; 36:12243-12258. [PMID: 27903732 DOI: 10.1523/jneurosci.1473-16.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 09/19/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022] Open
Abstract
Mutations in the gene encoding Parkin, an E3 ubiquitin ligase, lead to juvenile-onset Parkinson's disease by inducing the selective death of midbrain dopaminergic neurons. Accumulating evidence indicates that Parkin also has an important role in excitatory glutamatergic neurotransmission, although its precise mechanism of action remains unclear. Here, we investigate Parkin's role at glutamatergic synapses of rat hippocampal neurons. We find that Parkin-deficient neurons exhibit significantly reduced AMPA receptor (AMPAR)-mediated currents and cell-surface expression, and that these phenotypes result from decreased postsynaptic expression of the adaptor protein Homer1, which is necessary for coupling AMPAR endocytic zones with the postsynaptic density. Accordingly, Parkin loss of function leads to the reduced density of postsynaptic endocytic zones and to impaired AMPAR internalization. These findings demonstrate a novel and essential role for Parkin in glutamatergic neurotransmission, as a stabilizer of postsynaptic Homer1 and the Homer1-linked endocytic machinery necessary for maintaining normal cell-surface AMPAR levels. SIGNIFICANCE STATEMENT Mutations in Parkin, a ubiquitinating enzyme, lead to the selective loss of midbrain dopaminergic neurons and juvenile-onset Parkinson's disease (PD). Parkin loss of function has also been shown to alter hippocampal glutamatergic neurotransmission, providing a potential explanation for PD-associated cognitive impairment. However, very little is known about Parkin's specific sites or mechanisms of action at glutamatergic synapses. Here, we show that Parkin deficiency leads to decreased AMPA receptor-mediated activity due to disruption of the postsynaptic endocytic zones required for maintaining proper cell-surface AMPA receptor levels. These findings demonstrate a novel role for Parkin in synaptic AMPA receptor internalization and suggest a Parkin-dependent mechanism for hippocampal dysfunction that may explain cognitive deficits associated with some forms of PD.
Collapse
|
94
|
Calap-Quintana P, González-Fernández J, Sebastiá-Ortega N, Llorens JV, Moltó MD. Drosophila melanogaster Models of Metal-Related Human Diseases and Metal Toxicity. Int J Mol Sci 2017; 18:E1456. [PMID: 28684721 PMCID: PMC5535947 DOI: 10.3390/ijms18071456] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 12/21/2022] Open
Abstract
Iron, copper and zinc are transition metals essential for life because they are required in a multitude of biological processes. Organisms have evolved to acquire metals from nutrition and to maintain adequate levels of each metal to avoid damaging effects associated with its deficiency, excess or misplacement. Interestingly, the main components of metal homeostatic pathways are conserved, with many orthologues of the human metal-related genes having been identified and characterized in Drosophila melanogaster. Drosophila has gained appreciation as a useful model for studying human diseases, including those caused by mutations in pathways controlling cellular metal homeostasis. Flies have many advantages in the laboratory, such as a short life cycle, easy handling and inexpensive maintenance. Furthermore, they can be raised in a large number. In addition, flies are greatly appreciated because they offer a considerable number of genetic tools to address some of the unresolved questions concerning disease pathology, which in turn could contribute to our understanding of the metal metabolism and homeostasis. This review recapitulates the metabolism of the principal transition metals, namely iron, zinc and copper, in Drosophila and the utility of this organism as an experimental model to explore the role of metal dyshomeostasis in different human diseases. Finally, a summary of the contribution of Drosophila as a model for testing metal toxicity is provided.
Collapse
Affiliation(s)
- Pablo Calap-Quintana
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
| | - Javier González-Fernández
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain.
| | - Noelia Sebastiá-Ortega
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Spain.
| | - José Vicente Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
| | - María Dolores Moltó
- Department of Genetics, University of Valencia, Campus of Burjassot, 46100 Valencia, Spain.
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Spain.
| |
Collapse
|
95
|
Abstract
Nearly 20 years have passed since we identified the causative gene for a familial Parkinson's disease, parkin (now known as PARK2), in 1998. PARK2 is the most common gene responsible for young-onset Parkinson's disease. It codes for the protein Parkin RBR E3 ubiquitin-protein ligase (PARK2), which directly links to the ubiquitin-proteasome as a ubiquitin ligase. PARK2 is involved in mitophagy, which is a type of autophagy, in collaboration with PTEN-induced putative kinase 1 (PINK1). The PINK1 gene (previously known as PARK6) is also a causative gene for young-onset Parkinson's disease. Both gene products may be involved in regulating quality control within the mitochondria. The discovery of PARK2 as a cause of young-onset Parkinson's disease has had a major impact on other neurodegenerative diseases. The involvement of protein degradation systems has been implicated as a common mechanism for neurodegenerative diseases in which inclusion body formation is observed. The discovery of the involvement of PARK2 in Parkinson's disease focused attention on the involvement of protein degradation systems in neurodegenerative diseases. In this review, we focus on the history of the discovery of PARK2, the clinical phenotypes of patients with PARK2 mutations, and its functional roles.
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
| | - Yoshikuni Mizuno
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
96
|
Abstract
Oxidative stress has been implicated as a core contributor to the initiation and progression of multiple neurological diseases. Genetic and environmental factors can produce oxidative stress through mitochondrial dysfunction leading to the degeneration of dopaminergic and other neurons underlying Parkinson disease (PD). Although clinical trials of antioxidants have thus far failed to demonstrate slowed progression of PD, oxidative stress remains a compelling target. Rather than prompting abandonment of antioxidant strategies, these failures have raised the bar for justifying drug and dosing selections and for improving study designs to test for disease modification by antioxidants. Urate, the main antioxidant found in plasma as well as the end product of purine metabolism in humans, has emerged as a promising potential neuroprotectant with advantages that distinguish it from previously tested antioxidant agents. Uniquely, higher urate levels in plasma or cerebrospinal fluid (CSF) have been linked to both a lower risk of developing PD and to a slower rate of its subsequent progression in numerous large prospective epidemiological and clinical cohorts. Laboratory evidence that urate confers neuroprotection in cellular and animal models of PD, possibly via the Nrf2 antioxidant response pathway, further strengthened its candidacy for rapid clinical translation. An early phase trial of the urate precursor inosine demonstrated its capacity to safely produce well tolerated, long-term elevation of plasma and CSF urate in early PD, supporting a phase 3 trial now underway to determine whether oral inosine dosed to elevate urate to concentrations predictive of favorable prognosis in PD slows clinical decline in people with recently diagnosed, dopamine transporter-deficient PD.
Collapse
Affiliation(s)
- Grace F Crotty
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Alberto Ascherio
- Departments of Epidemiology and Nutrition, Harvard School of Public Health, Boston, MA, USA
| | | |
Collapse
|
97
|
Kim T, Vemuganti R. Mechanisms of Parkinson's disease-related proteins in mediating secondary brain damage after cerebral ischemia. J Cereb Blood Flow Metab 2017; 37:1910-1926. [PMID: 28273718 PMCID: PMC5444552 DOI: 10.1177/0271678x17694186] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Both Parkinson's disease (PD) and stroke are debilitating conditions that result in neuronal death and loss of neurological functions. These two conditions predominantly affect aging populations with the deterioration of the quality of life for the patients themselves and a tremendous burden to families. While the neurodegeneration and symptomology of PD develop chronically over the years, post-stroke neuronal death and dysfunction develop rapidly in days. Despite the discrepancy in the pathophysiological time frame and severity, both conditions share common molecular mechanisms that include oxidative stress, mitochondrial dysfunction, inflammation, endoplasmic reticulum stress, and activation of various cell death pathways (apoptosis/necrosis/autophagy) that synergistically modulate the neuronal death. Emerging evidence indicates that several proteins associated with early-onset familial PD play critical roles in mediating the neuronal death. Importantly, mutations in the genes encoding Parkin, PTEN-induced putative kinase 1 and DJ-1 mediate autosomal recessive forms of PD, whereas mutations in the genes encoding leucine-rich repeat kinase 2 and α-synuclein are responsible for autosomal dominant PD. This review discusses the significance of these proteins with the emphasis on the role of α-synuclein in mediating post-ischemic brain damage.
Collapse
Affiliation(s)
- TaeHee Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,2 Neuroscience Training Program, Madison, WI, USA
| | - Raghu Vemuganti
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,2 Neuroscience Training Program, Madison, WI, USA.,3 Cellular & Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.,4 William S. Middleton Memorial Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
98
|
Abstract
Since the first formal description of Parkinson disease (PD) two centuries ago, our understanding of this common neurodegenerative disorder has expanded at all levels of description, from the delineation of its clinical phenotype to the identification of its neuropathological features, neurochemical processes and genetic factors. Along the way, findings have led to novel hypotheses about how the disease develops and progresses, challenging our understanding of how neurodegenerative disorders wreak havoc on human health. In this Timeline article, I recount the fascinating 200-year journey of PD research.
Collapse
Affiliation(s)
- Serge Przedborski
- Departments of Neurology, Pathology, and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| |
Collapse
|
99
|
Park MH, Lee HJ, Lee HL, Son DJ, Ju JH, Hyun BK, Jung SH, Song JK, Lee DH, Hwang CJ, Han SB, Kim S, Hong JT. Parkin Knockout Inhibits Neuronal Development via Regulation of Proteasomal Degradation of p21. Am J Cancer Res 2017; 7:2033-2045. [PMID: 28656059 PMCID: PMC5485421 DOI: 10.7150/thno.19824] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 03/31/2017] [Indexed: 11/05/2022] Open
Abstract
PARK2 encodes for the E3 ubiquitin ligase parkin and is implicated in the development of Parkinson's disease (PD). Although the neuroprotective role of parkin is well known, the mechanism of PARK2's function in neural stem differentiation has not yet been thoroughly studied. Co-expressions network analysis showed that synaptosomal-associated protein 25 (SNAP-25) and brain-derived neurotrophic factor (BDNF) were positively correlated with parkin, but negatively correlated with p21 in human patient brain. We investigated a link between the ubiquitin E3 ligase parkin and proteasomal degradation of p21 for the control of neural stem cell differentiation. We found that the neurogenesis was lowered in PARK2 knockout (KO) mice compared with non-tg mice. Expression of the marker protein for neural cell differentiation such as class III beta tubulin (TUBBIII), glial fibrillary acidic protein (GFAP) and neurofilament, as well as SNAP25 and BDNF, was down regulated in PARK2 KO mice. Associated with the loss of differentiation function, p21 protein was highly accumulated in the neural stem cells of PARK2 KO mice. We discovered that p21 directly binds with parkin and is ubiquitinated by parkin which resulted in the loss of cell differentiation ability. Introduction of p21 shRNA in PARK2 KO mice significantly rescued the differentiation efficacy as well as SNAP25 and BDNF expression. c-Jun N-terminal kinase (JNK) pathway is implicated in neurogenesis and p21 degradation. We also defined the decreased p21 ubiquitination and differentiation ability were reversed after treatment with JNK inhibitor, SP600125 in PARK2 KO mice derived neural stem cells. Thus, the present study indicated that parkin knockout inhibits neural stem cell differentiation by JNK-dependent proteasomal degradation of p21.
Collapse
|
100
|
Buneeva OA, Medvedev AE. The role of atypical ubiquitination in cell regulation. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2017. [DOI: 10.1134/s1990750817010024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|