51
|
Alpha-Synuclein PET Tracer Development-An Overview about Current Efforts. Pharmaceuticals (Basel) 2021; 14:ph14090847. [PMID: 34577548 PMCID: PMC8466155 DOI: 10.3390/ph14090847] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases such as Parkinson’s disease (PD) are manifested by inclusion bodies of alpha-synuclein (α-syn) also called α-synucleinopathies. Detection of these inclusions is thus far only possible by histological examination of postmortem brain tissue. The possibility of non-invasively detecting α-syn will therefore provide valuable insights into the disease progression of α-synucleinopathies. In particular, α-syn imaging can quantify changes in monomeric, oligomeric, and fibrillic α-syn over time and improve early diagnosis of various α-synucleinopathies or monitor treatment progress. Positron emission tomography (PET) is a non-invasive in vivo imaging technique that can quantify target expression and drug occupancies when a suitable tracer exists. As such, novel α-syn PET tracers are highly sought after. The development of an α-syn PET tracer faces several challenges. For example, the low abundance of α-syn within the brain necessitates the development of a high-affinity ligand. Moreover, α-syn depositions are, in contrast to amyloid proteins, predominantly localized intracellularly, limiting their accessibility. Furthermore, another challenge is the ligand selectivity over structurally similar amyloids such as amyloid-beta or tau, which are often co-localized with α-syn pathology. The lack of a defined crystal structure of α-syn has also hindered rational drug and tracer design efforts. Our objective for this review is to provide a comprehensive overview of current efforts in the development of selective α-syn PET tracers.
Collapse
|
52
|
Dhanawat M, Mehta DK, Gupta S, Das R. Understanding the Pathogenesis Involved in Parkinson's Disease and Potential Therapeutic Treatment Strategies. Cent Nerv Syst Agents Med Chem 2021; 20:88-102. [PMID: 32628600 DOI: 10.2174/1871524920666200705222842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 01/01/2023]
Abstract
A vast advancement has been made in the treatment related to central nervous system disorders especially Parkinson's disease. The development in therapeutics and a better understanding of the targets results in upsurge of many promising therapies for Parkinson's disease. Parkinson's disease is defined by neuronal degeneration and neuroinflammation and it is reported that the presence of the neurofibrillary aggregates such as Lewy bodies is considered as the marker. Along with this, it is also characterized by the presence of motor and non-motor symptoms, as seen in Parkinsonian patients. A lot of treatment options mainly focus on prophylactic measures or the symptomatic treatment of Parkinson's disease. Neuroinflammation and neurodegeneration are the point of interest which can be exploited as a new target to emphasis on Parkinson's disease. A thorough study of these targets helps in modifications of those molecules which are particularly involved in causing the neuronal degeneration and neuroinflammation in Parkinson's disease. A lot of drug regimens are available for the treatment of Parkinson's disease, although levodopa remains the choice of drug for controlling the symptoms, yet is accompanied with significant snags. It is always suggested to use other drug therapies concomitantly with levodopa. A number of significant causes and therapeutic targets for Parkinson's disease have been identified in the last decade, here an attempt was made to highlight the most significant of them. It was also found that the treatment regimen and involvement of therapies are totally dependent on individuals and can be tailored to the needs of each individual patient.
Collapse
Affiliation(s)
- Meenakshi Dhanawat
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, HR-133207, India
| | - Dinesh K Mehta
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, HR-133207, India
| | - Sumeet Gupta
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, HR-133207, India
| | - Rina Das
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, HR-133207, India
| |
Collapse
|
53
|
Deng I, Corrigan F, Garg S, Zhou XF, Bobrovskaya L. Further Characterization of Intrastriatal Lipopolysaccharide Model of Parkinson's Disease in C57BL/6 Mice. Int J Mol Sci 2021; 22:7380. [PMID: 34299000 PMCID: PMC8304722 DOI: 10.3390/ijms22147380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is the most common movement disorder, characterized by progressive degeneration of the nigrostriatal pathway, which consists of dopaminergic cell bodies in substantia nigra and their neuronal projections to the striatum. Moreover, PD is associated with an array of non-motor symptoms such as olfactory dysfunction, gastrointestinal dysfunction, impaired regulation of the sleep-wake cycle, anxiety, depression, and cognitive impairment. Inflammation and concomitant oxidative stress are crucial in the pathogenesis of PD. Thus, this study aimed to model PD via intrastriatal injection of the inflammagen lipopolysaccharide (LPS)to investigate if the lesion causes olfactory and motor impairments, inflammation, oxidative stress, and alteration in synaptic proteins in the olfactory bulb, striatum, and colon. Ten µg of LPS was injected unilaterally into the striatum of 27 male C57BL/6 mice, and behavioural assessment was conducted at 4 and 8 weeks post-treatment, followed by tissue collection. Intrastriatal LPS induced motor impairment in C57BL/6 mice at 8 weeks post-treatment evidenced by reduced latency time in the rotarod test. LPS also induced inflammation in the striatum characterized by increased expression of microglial marker Iba-1 and astrocytic marker GFAP, with degeneration of dopaminergic neuronal fibres (reduced tyrosine hydroxylase immunoreactivity), and reduction of synaptic proteins and DJ-1 protein. Additionally, intrastriatal LPS induced inflammation, oxidative stress and alterations in synaptic proteins within the olfactory bulb, although this did not induce a significant impairment in olfactory function. Intrastriatal LPS induced mild inflammatory changes in the distal colon, accompanied by increased protein expression of 3-nitrotyrosine-modified proteins. This model recapitulated the major features of PD such as motor impairment and degeneration of dopaminergic neuronal fibres in the striatum, as well as some pathological changes in the olfactory bulb and colon; thus, this model could be suitable for understanding clinical PD and testing neuroprotective strategies.
Collapse
Affiliation(s)
- Isaac Deng
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (I.D.); (S.G.); (X.-F.Z.)
| | - Frances Corrigan
- Medical Sciences, University of Adelaide, Adelaide 5000, Australia;
| | - Sanjay Garg
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (I.D.); (S.G.); (X.-F.Z.)
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (I.D.); (S.G.); (X.-F.Z.)
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia; (I.D.); (S.G.); (X.-F.Z.)
| |
Collapse
|
54
|
Gaggi G, Di Credico A, Izzicupo P, Iannetti G, Di Baldassarre A, Ghinassi B. Chemical and Biological Molecules Involved in Differentiation, Maturation, and Survival of Dopaminergic Neurons in Health and Parkinson's Disease: Physiological Aspects and Clinical Implications. Biomedicines 2021; 9:biomedicines9070754. [PMID: 34209807 PMCID: PMC8301385 DOI: 10.3390/biomedicines9070754] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disease characterized by a specific and progressive loss of dopaminergic (DA) neurons and dopamine, causing motor dysfunctions and impaired movements. Unfortunately, available therapies can partially treat the motor symptoms, but they have no effect on non-motor features. In addition, the therapeutic effect reduces gradually, and the prolonged use of drugs leads to a significative increase in the number of adverse events. For these reasons, an alternative approach that allows the replacement or the improved survival of DA neurons is very appealing for the treatment of PD patients and recently the first human clinical trials for DA neurons replacement have been set up. Here, we review the role of chemical and biological molecules that are involved in the development, survival and differentiation of DA neurons. In particular, we review the chemical small molecules used to differentiate different type of stem cells into DA neurons with high efficiency; the role of microRNAs and long non-coding RNAs both in DA neurons development/survival as far as in the pathogenesis of PD; and, finally, we dissect the potential role of exosomes carrying biological molecules as treatment of PD.
Collapse
Affiliation(s)
- Giulia Gaggi
- Beth Israel Deaconess Medical Center, Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA;
| | - Andrea Di Credico
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| | - Pascal Izzicupo
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| | | | - Angela Di Baldassarre
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
- Correspondence:
| | - Barbara Ghinassi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| |
Collapse
|
55
|
Teng JS, Ooi YY, Chye SM, Ling APK, Koh RY. Immunotherapies for Parkinson's disease: Progression of Clinical Development. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:802-813. [PMID: 34042040 DOI: 10.2174/1871527320666210526160926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease is a common neurodegenerative disease affecting the movement and wellbeing of most elderlies. The manifestations of Parkinson's disease often include resting tremor, stiffness, bradykinesia and muscular rigidity. The typical hallmark of Parkinson's disease is the destruction of neurons in the substantia nigra and the presence of Lewy bodies in different compartments of the central nervous system. Due to various limitations to the currently available treatments, immunotherapies have emerged to be the new approach to Parkinson's disease treatment. This approach shows some positive outcomes on the efficacy in removing the aggregated species of alpha-synuclein, which is believed to be one of the causes of Parkinson's disease. In this review, an overview of how alpha-synuclein contributes to Parkinson's disease and the effects of a few new immunotherapeutic treatments, including BIIB054 (cinpanemab), MEDI1341, AFFITOPE and PRX002 (prasinezumab) that are currently under clinical development, will be discussed.
Collapse
Affiliation(s)
- Jet Shee Teng
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| | - Yin Yin Ooi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, Selangor, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
56
|
Oganesyan I, Lento C, Tandon A, Wilson DJ. Conformational Dynamics of α-Synuclein during the Interaction with Phospholipid Nanodiscs by Millisecond Hydrogen-Deuterium Exchange Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1169-1179. [PMID: 33784451 DOI: 10.1021/jasms.0c00463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Both normal and pathological functions of α-synuclein (αSN), an abundant protein in the central and peripheral nervous system, have been linked to its interaction with membrane lipid bilayers. The ability to characterize structural transitions of αSN upon membrane complexation will clarify molecular mechanisms associated with αSN-linked pathologies, including Parkinson's disease (PD), multiple systems atrophy, and other synucleinopathies. In this work, time-resolved electrospray ionization hydrogen/deuterium exchange mass spectrometry (TRESI-HDX-MS) was employed to acquire a detailed picture of αSN's conformational transitions as it undergoes complexation with nanodisc membrane mimics with different headgroup charges (zwitterionic DMPC and negative POPG). Using this approach, αSN interactions with DMPC nanodiscs were shown to be rapid exchanging and to have little impact on the αSN conformational ensemble. Interactions with nanodiscs containing lipids known to promote amyloidogenesis (e.g., POPG), on the other hand, were observed to induce substantial and specific changes in the αSN conformational ensemble. Ultimately, we identify a region corresponding residues 19-28 and 45-57 of the αSN sequence that is uniquely impacted by interactions with "amyloidogenic" lipid membranes, supporting the existing "broken-helix" model for α-synuclein/membrane interactions, but do not detect a "helical extension" that is also thought to play a role in αSN aggregation.
Collapse
Affiliation(s)
- Irina Oganesyan
- Department of Chemistry, York University, Toronto M3J 1P3, Canada
| | - Cristina Lento
- Department of Chemistry, York University, Toronto M3J 1P3, Canada
| | - Anurag Tandon
- Department of Medicine, University of Toronto, Toronto M5S 1A1, Canada
| | - Derek J Wilson
- Department of Chemistry, York University, Toronto M3J 1P3, Canada
- Centre for Research in Mass Spectrometry, York University, Toronto M3J 1P3, Canada
| |
Collapse
|
57
|
Prasuhn J, Brüggemann N. Genotype-driven therapeutic developments in Parkinson's disease. Mol Med 2021; 27:42. [PMID: 33874883 PMCID: PMC8056568 DOI: 10.1186/s10020-021-00281-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Remarkable advances have been reached in the understanding of the genetic basis of Parkinson's disease (PD), with the identification of monogenic causes (mPD) and a plethora of gene loci leading to an increased risk for idiopathic PD. The expanding knowledge and subsequent identification of genetic contributions fosters the understanding of molecular mechanisms leading to disease development and progression. Distinct pathways involved in mitochondrial dysfunction, oxidative stress, and lysosomal function have been identified and open a unique window of opportunity for individualized treatment approaches. These genetic findings have led to an imminent progress towards pathophysiology-targeted clinical trials and potentially disease-modifying treatments in the future. MAIN BODY OF THE MANUSCRIPT In this review article we will summarize known genetic contributors to the pathophysiology of Parkinson's disease, the molecular mechanisms leading to disease development, and discuss challenges and opportunities in clinical trial designs. CONCLUSIONS The future success of clinical trials in PD is mainly dependent on reliable biomarker development and extensive genetic testing to identify genetic cases. Whether genotype-dependent stratification of study participants will extend the potential application of new drugs will be one major challenge in conceptualizing clinical trials. However, the latest developments in genotype-driven treatments will pave the road to individualized pathophysiology-based therapies in the future.
Collapse
Affiliation(s)
- Jannik Prasuhn
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
58
|
High-resolution ultrasound changes of the vagus nerve in idiopathic Parkinson's disease (IPD): a possible additional index of disease. Neurol Sci 2021; 42:5205-5211. [PMID: 33821361 PMCID: PMC8642255 DOI: 10.1007/s10072-021-05183-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/13/2021] [Indexed: 12/13/2022]
Abstract
Background and rationale Histopathological studies revealed degeneration of the dorsal motor nucleus of the vagus nerve (VN) early in the course of idiopathic Parkinson’s disease (IPD). Degeneration of VN axons should be detectable by high-resolution ultrasound (HRUS) as a thinning of the nerve trunk. In order to establish if the VN exhibits sonographic signs of atrophy in IPD, we examined patients with IPD compared with age-matched controls. Material and methods We measured the caliber (cross-sectional area, CSA) and perimeter of the VN in 20 outpatients with IPD (8 females and 12 males; mean age 73.0 + 8.6 years) and in age-matched controls using HRUS. Evaluation was performed by blinded raters using an Esaote MyLab Gamma device in conventional B-Mode with an 8–19 MHz probe. Results In both sides, the VN CSA was significantly smaller in IPD outpatients than in controls (right 2.37 + 0.91, left 1.87 + 1.35 mm2 versus 6.0 + 1.33, 5.6 + 1.26 mm2; p <0.001), as well as the perimeter (right 5.06 + 0.85, left 4.78 + 1.74 mm versus 8.87 + 0.86, 8.58 + 0.97 mm; p <0.001). There were no significant correlations between VN CSA and age, the Hoehn and Yahr scale, L-dopa therapy, and disease duration. Conclusion Our findings provide evidence of atrophy of the VNs in IPD patients by HRUS. Moreover, HRUS of the VN represent a non-invasive easy imaging modality of screening in IPD patients independent of disease stage and duration and an interesting possible additional index of disease.
Collapse
|
59
|
Ullah I, Zhao L, Hai Y, Fahim M, Alwayli D, Wang X, Li H. "Metal elements and pesticides as risk factors for Parkinson's disease - A review". Toxicol Rep 2021; 8:607-616. [PMID: 33816123 PMCID: PMC8010213 DOI: 10.1016/j.toxrep.2021.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Essential metals including iron (Fe) and manganese (Mn) with known physiological functions in human body play an important role in cell homeostasis. Excessive exposure to these essential as well as non-essential metals including mercury (Hg) and Aluminum (Al) may contribute to pathological conditions, including PD. Each metal could be toxic through specific pathways. Epidemiological evidences from occupational and ecological studies besides various in vivo and in vitro studies have revealed the possible pathogenic role and neurotoxicity of different metals. Pesticides are substances that aim to mitigate the harm done by pests to plants and crops, and are extensively used to boost agricultural production. This review provides an outline of our current knowledge on the possible association between metals and PD. We have discussed the potential association between these two, furthermore the chemical properties, biological and toxicological aspects as well as possible mechanisms of Fe, Mn, Cu, Zn, Al, Ca, Pb, Hg and Zn in PD pathogenesis. In addition, we review recent evidence on deregulated microRNAs upon pesticide exposure and possible role of deregulated miRNA and pesticides to PD pathogenesis.
Collapse
Affiliation(s)
- Inam Ullah
- School of Life Sciences, Lanzhou University, China
| | - Longhe Zhao
- School of Pharmacy, Lanzhou University, China
| | - Yang Hai
- School of Pharmacy, Lanzhou University, China
| | | | | | - Xin Wang
- School of Pharmacy, Lanzhou University, China
| | - Hongyu Li
- School of Life Sciences, Lanzhou University, China
- School of Pharmacy, Lanzhou University, China
| |
Collapse
|
60
|
Suppressing Cdk5 Activity by Luteolin Inhibits MPP +-Induced Apoptotic of Neuroblastoma through Erk/Drp1 and Fak/Akt/GSK3β Pathways. Molecules 2021; 26:molecules26051307. [PMID: 33671094 PMCID: PMC7957557 DOI: 10.3390/molecules26051307] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by the progressive degeneration of dopaminergic neurons. The cause of PD is still unclear. Oxidative stress and mitochondrial dysfunction have been linked to the development of PD. Luteolin, a non-toxic flavonoid, has become interested in an alternative medicine, according to its effects on anti-oxidative stress and anti-apoptosis, although the underlying mechanism of luteolin on PD has not been fully elucidated. This study aims to investigate whether luteolin prevents neurotoxicity induction by 1-methyl-4-phenylpyridinium iodide (MPP+), a neurotoxin in neuroblastoma SH-SY5Y cells. The results reveal that luteolin significantly improved cell viability and reduced apoptosis in MPP+-treated cells. Increasing lipid peroxidation and superoxide anion (O2−), including mitochondrial membrane potential (Δψm) disruption, is ameliorated by luteolin treatment. In addition, luteolin attenuated MPP+-induced neurite damage via GAP43 and synapsin-1. Furthermore, Cdk5 is found to be overactivated and correlated with elevation of cleaved caspase-3 activity in MPP+-exposed cells, while phosphorylation of Erk1/2, Drp1, Fak, Akt and GSK3β are inhibited. In contrast, luteolin attenuated Cdk5 overactivation and supported phosphorylated level of Erk1/2, Drp1, Fak, Akt and GSK3β with reducing in cleaved caspase-3 activity. Results indicate that luteolin exerts neuroprotective effects via Cdk5-mediated Erk1/2/Drp1 and Fak/Akt/GSK3β pathways, possibly representing a potential preventive agent for neuronal disorder.
Collapse
|
61
|
Liu H, Koros C, Strohäker T, Schulte C, Bozi M, Varvaresos S, Ibáñez de Opakua A, Simitsi AM, Bougea A, Voumvourakis K, Maniati M, Papageorgiou SG, Hauser AK, Becker S, Zweckstetter M, Stefanis L, Gasser T. A Novel SNCA A30G Mutation Causes Familial Parkinson's Disease. Mov Disord 2021; 36:1624-1633. [PMID: 33617693 DOI: 10.1002/mds.28534] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/15/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The SNCA gene encoding α-synuclein (αSyn) is the first gene identified to cause autosomal-dominant Parkinson's disease (PD). OBJECTIVE We report the identification of a novel heterozygous A30G mutation of the SNCA gene in familial PD and describe clinical features of affected patients, genetic findings, and functional consequences. METHODS Whole exome sequencing was performed in the discovery family proband. Restriction digestion with Bbvl was used to screen SNCA A30G in two validation cohorts. The Greek cohort included 177 familial PD probands, 109 sporadic PD cases, and 377 neurologically healthy controls. The German cohort included 136 familial PD probands, 380 sporadic PD cases, and 116 neurologically healthy controls. We also conducted haplotype analysis using 13 common single nucleotide variants around A30G to determine the possibility of a founder effect for A30G. We then used biophysical methods to characterize A30G αSyn. RESULTS We identified a novel SNCA A30G (GRCh37, Chr4:90756730, c.89 C>G) mutation that co-segregated with the disease in five affected individuals of three Greek families and was absent from controls. A founder effect was strongly suggested by haplotype analysis. The A30G mutation had a local effect on the intrinsically disordered structure of αSyn, slightly perturbed membrane binding, and promoted fibril formation. CONCLUSION Based on the identification of A30G co-segregating with the disease in three families, the absence of the mutation in controls and population databases, and the observed functional effects, we propose SNCA A30G as a novel causative mutation for familial PD. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Hui Liu
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen and German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Christos Koros
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece.,2nd Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Timo Strohäker
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Claudia Schulte
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen and German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Maria Bozi
- 2nd Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefanos Varvaresos
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Athina Maria Simitsi
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Bougea
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Voumvourakis
- 2nd Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Matina Maniati
- Laboratory of Neurodegenerative Diseases, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sokratis G Papageorgiou
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece.,2nd Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ann-Kathrin Hauser
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen and German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Stefan Becker
- Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Leonidas Stefanis
- 1st Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece.,Laboratory of Neurodegenerative Diseases, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Thomas Gasser
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen and German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| |
Collapse
|
62
|
Frataxins Emerge as New Players of the Intracellular Antioxidant Machinery. Antioxidants (Basel) 2021; 10:antiox10020315. [PMID: 33672495 PMCID: PMC7923443 DOI: 10.3390/antiox10020315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022] Open
Abstract
Frataxin is a mitochondrial protein which deficiency causes Friedreich's ataxia, a cardio-neurodegenerative disease. The lack of frataxin induces the dysregulation of mitochondrial iron homeostasis and oxidative stress, which finally causes the neuronal death. The mechanism through which frataxin regulates the oxidative stress balance is rather complex and poorly understood. While the absence of human (Hfra) and yeast (Yfh1) frataxins turn out cells sensitive to oxidative stress, this does not occur when the frataxin gene is knocked-out in E. coli. To better understand the biological roles of Hfra and Yfh1 as endogenous antioxidants, we have studied their ability to inhibit the formation of reactive oxygen species (ROS) from Cu2+- and Fe3+-catalyzed degradation of ascorbic acid. Both proteins drastically reduce the formation of ROS, and during this process they are not oxidized. In addition, we have also demonstrated that merely the presence of Yfh1 or Hfra is enough to protect a highly oxidation-prone protein such as α-synuclein. This unspecific intervention (without a direct binding) suggests that frataxins could act as a shield to prevent the oxidation of a broad set of intracellular proteins, and reinforces that idea that frataxin can be used to prevent neurological pathologies linked to an enhanced oxidative stress.
Collapse
|
63
|
Jin JW, Fan X, Del Cid-Pellitero E, Liu XX, Zhou L, Dai C, Gibbs E, He W, Li H, Wu X, Hill A, Leavitt BR, Cashman N, Liu L, Lu J, Durcan TM, Dong Z, Fon EA, Wang YT. Development of an α-synuclein knockdown peptide and evaluation of its efficacy in Parkinson's disease models. Commun Biol 2021; 4:232. [PMID: 33608634 PMCID: PMC7895943 DOI: 10.1038/s42003-021-01746-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 12/16/2020] [Indexed: 11/14/2022] Open
Abstract
Convincing evidence supports the premise that reducing α-synuclein levels may be an effective therapy for Parkinson's disease (PD); however, there has been lack of a clinically applicable α-synuclein reducing therapeutic strategy. This study was undertaken to develop a blood-brain barrier and plasma membrane-permeable α-synuclein knockdown peptide, Tat-βsyn-degron, that may have therapeutic potential. The peptide effectively reduced the level of α-synuclein via proteasomal degradation both in cell cultures and in animals. Tat-βsyn-degron decreased α-synuclein aggregates and microglial activation in an α-synuclein pre-formed fibril model of spreading synucleinopathy in transgenic mice overexpressing human A53T α-synuclein. Moreover, Tat-βsyn-degron reduced α-synuclein levels and significantly decreased the parkinsonian toxin-induced neuronal damage and motor impairment in a mouse toxicity model of PD. These results show the promising efficacy of Tat-βsyn-degron in two different animal models of PD and suggest its potential use as an effective PD therapeutic that directly targets the disease-causing process.
Collapse
Affiliation(s)
- Jack Wuyang Jin
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Xuelai Fan
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Esther Del Cid-Pellitero
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Xing-Xing Liu
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Limin Zhou
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chunfang Dai
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ebrima Gibbs
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wenting He
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongjie Li
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobin Wu
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Austin Hill
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Neil Cashman
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lidong Liu
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jie Lu
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Thomas M Durcan
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Edward A Fon
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Yu Tian Wang
- The Djavad Mowafaghian Centre for Brain Health and Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
64
|
Interaction between Parkin and α-Synuclein in PARK2-Mediated Parkinson's Disease. Cells 2021; 10:cells10020283. [PMID: 33572534 PMCID: PMC7911026 DOI: 10.3390/cells10020283] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Parkin and α-synuclein are two key proteins involved in the pathophysiology of Parkinson's disease (PD). Neurotoxic alterations of α-synuclein that lead to the formation of toxic oligomers and fibrils contribute to PD through synaptic dysfunction, mitochondrial impairment, defective endoplasmic reticulum and Golgi function, and nuclear dysfunction. In half of the cases, the recessively inherited early-onset PD is caused by loss of function mutations in the PARK2 gene that encodes the E3-ubiquitin ligase, parkin. Parkin is involved in the clearance of misfolded and aggregated proteins by the ubiquitin-proteasome system and regulates mitophagy and mitochondrial biogenesis. PARK2-related PD is generally thought not to be associated with Lewy body formation although it is a neuropathological hallmark of PD. In this review article, we provide an overview of post-mortem neuropathological examinations of PARK2 patients and present the current knowledge of a functional interaction between parkin and α-synuclein in the regulation of protein aggregates including Lewy bodies. Furthermore, we describe prevailing hypotheses about the formation of intracellular micro-aggregates (synuclein inclusions) that might be more likely than Lewy bodies to occur in PARK2-related PD. This information may inform future studies aiming to unveil primary signaling processes involved in PD and related neurodegenerative disorders.
Collapse
|
65
|
Li Q, Wang Z, Xing H, Wang Y, Guo Y. Exosomes derived from miR-188-3p-modified adipose-derived mesenchymal stem cells protect Parkinson's disease. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 23:1334-1344. [PMID: 33717653 PMCID: PMC7920810 DOI: 10.1016/j.omtn.2021.01.022] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 01/17/2021] [Indexed: 12/19/2022]
Abstract
Parkinson’s disease (PD) is the second-most common neurodegenerative disease after Alzheimer’s disease. The most important pathological feature of PD is the irreversible damage of dopamine neurons, which is related to autophagy and neuroinflammation in the substantia nigra. Previous studies found that the activation of NAcht Leucine-rich repeat Protein 3 (NLRP3) inflammasome/pyroptosis and cell division protein kinase 5 (CDK5)-mediated autophagy played an important role in PD. Bioinformatics analyses further predicted that microRNA (miR)-188-3p potentially targets NLRP3 and CDK5. Adipose-derived stem cell (ADSC)-derived exosomes were found to be excellent vectors for genetic therapy. We assessed the levels of injury, autophagy, and inflammasomes in 1-methyl-4-phenyl-1,2,4,5-tetrahydropyridine (MPTP)-induced PD mice models and neurotoxin 1-methyl-4-phenylpyridinium (MPP+)-induced cell models after treating them with miR-188-3p-enriched exosomes. miR-188-3p-enriched exosome treatment suppressed autophagy and pyroptosis, whereas increased proliferation via targeting CDK5 and NLRP3 in mice and MN9D cells. It was revealed that mir-188-3p could be a new therapeutic target for curing PD patients.
Collapse
Affiliation(s)
- Qiang Li
- The First Affiliated Hospital of Bengbu Medical College, Anhui, China
| | - Zihao Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Guo
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
66
|
Han NR, Kim YK, Ahn S, Hwang TY, Lee H, Park HJ. A Comprehensive Phenotype of Non-motor Impairments and Distribution of Alpha-Synuclein Deposition in Parkinsonism-Induced Mice by a Combination Injection of MPTP and Probenecid. Front Aging Neurosci 2021; 12:599045. [PMID: 33519420 PMCID: PMC7838388 DOI: 10.3389/fnagi.2020.599045] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is characterized by non-motor symptoms as well as motor deficits. The non-motor symptoms rarely appear individually and occur simultaneously with motor deficits or independently. However, a comprehensive research on the non-motor symptoms using an experimental model of PD remains poorly understood. The aim of the current study is to establish a chronic mouse model of PD mimicking the comprehensive non-motor symptoms of human PD by injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and probenecid (MPTP/p). The non-motor and motor symptoms were evaluated by performing buried food, short-term olfactory memory, hot plate, open field, tail suspension, Y maze, novel object recognition, bead expulsion, one-h stool collection, rotarod, rearing, catalepsy, and akinesia tests after 10 injections of MPTP/p into mice. The expression levels of α-synuclein, glial fibrillary acidic protein (GFAP), tyrosine hydroxylase (TH) or DJ-1 were analyzed by Western blotting or immunostaining. MPTP/p-treated mice achieved to reproduce the key features of non-motor symptoms including olfactory deficit, thermal hyperalgesia, anxiety, depression, cognitive decline, and gastrointestinal dysfunction in addition to motor deficits. The MPTP/p-treated mice also showed the high levels of α-synuclein and low levels of TH and DJ-1 in striatum, substantia nigra, olfactory bulb, hippocampus, amygdala, prefrontal cortex, locus coeruleus, or colon. In addition, the expression levels of phosphorylated-α-synuclein and GFAP were elevated in the striatum and substantia nigra in the MPTP/p-treated mice. Taken together, our study clarifies that the chronic MPTP/p-treated mice have a variety of non-motor dysfunctions as well as motor abnormalities by α-synuclein overexpression and dopaminergic depletion. Therefore, the study of comprehensive phenotypes of non-motor symptoms in one PD model would advance in-depth understandings of neuropathological alternations and contribute to future strategies for PD treatment.
Collapse
Affiliation(s)
- Na-Ra Han
- Integrative Parkinson's Disease Research Group, Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, South Korea.,Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Yu-Kang Kim
- Integrative Parkinson's Disease Research Group, Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, South Korea.,Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Sora Ahn
- Integrative Parkinson's Disease Research Group, Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, South Korea
| | - Tae-Yeon Hwang
- Integrative Parkinson's Disease Research Group, Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, South Korea.,Department of Meridian & Acupoints, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyejung Lee
- Department of Meridian & Acupoints, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hi-Joon Park
- Integrative Parkinson's Disease Research Group, Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, South Korea.,Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
67
|
Zamanian M, Bazmandegan G, Sureda A, Sobarzo-Sanchez E, Yousefi-Manesh H, Shirooie S. The Protective Roles and Molecular Mechanisms of Troxerutin (Vitamin P4) for the Treatment of Chronic Diseases: A Mechanistic Review. Curr Neuropharmacol 2020; 19:97-110. [PMID: 32386493 PMCID: PMC7903491 DOI: 10.2174/1570159x18666200510020744] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/21/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
Troxerutin (TRX), a semi-synthetic bioflavonoid derived from rutin, has been reported to exert several pharmacological effects including antioxidant, anti-inflammatory, antihyperlipidemic, and nephroprotective. However, the related molecular details and its mechanisms remain poorly understood. In the present review, we presented evidences from the diversity in vitro and in vivo studies on the therapeutic potential of TRX against neurodegenerative, diabetes, cancer and cardiovascular diseases with the purpose to find molecular pathways related to the treatment efficacy. TRX has a beneficial role in many diseases through multiple mechanisms including, increasing antioxidant enzymes and reducing oxidative damage, decreasing in proapoptotic proteins (APAF-1, BAX, caspases-9 and-3) and increasing the antiapoptotic BCL-2, increasing the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and downregulating the nuclear factor κB (NFκ). TRX also reduces acetylcholinesterase activity and upregulates phosphoinositide 3- kinase/Akt signaling pathway in Alzheimer's disease models. Natural products such as TRX may develop numerous and intracellular pathways at several steps in the treatment of many diseases. Molecular mechanisms of action are revealing novel, possible combinational beneficial approaches to treat multiple pathological conditions.
Collapse
Affiliation(s)
| | - Gholamreza Bazmandegan
- Clinical Research Development Unit, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX), Health Research Institute of the Balearic Islands (IdISBa) and CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), University of Balearic Islands, Palma de Mallorca E-07122, Balearic Islands, Spain
| | - Eduardo Sobarzo-Sanchez
- Instituto de Investigación e Innovación en Salud, Facultyad de Ciencias de la Salud, Universidad Central de Chile, Chile
| | - Hasan Yousefi-Manesh
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, 13145-784, Tehran, Iran
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
68
|
Kang J, Kim JW, Heo H, Lee J, Park KY, Yoon JH, Chang J. Identification of BAG2 and Cathepsin D as Plasma Biomarkers for Parkinson's Disease. Clin Transl Sci 2020; 14:606-616. [PMID: 33202088 PMCID: PMC7993325 DOI: 10.1111/cts.12920] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/10/2020] [Indexed: 12/30/2022] Open
Abstract
The current diagnosis of Parkinson’s disease (PD) mostly relies on clinical rating scales related to motor dysfunction. Given that clinical symptoms of PD appear after significant neuronal cell death in the brain, it is required to identify accessible, objective, and quantifiable biomarkers for early diagnosis of PD. In this study, a total of 20 patients with idiopathic PD and 20 age‐matched patients with essential tremor according to the UK Brain Bank Criteria were consecutively enrolled to identify peripheral blood biomarkers for PD. Clinical data were obtained by clinical survey and assessment. Using albumin‐depleted and immunoglobulin G‐depleted plasma samples, we performed immunoblot analysis of seven autophagy‐related proteins and compared the levels of proteins to those of the control group. We also analyzed the correlation between the levels of candidate proteins and clinical characteristics. Finally, we validated our biomarker models using receiver operating characteristic curve analysis. We found that the levels of BCL2‐associated athanogene 2 (BAG2) and cathepsin D were significantly decreased in plasma of patients with PD (P = 0.009 and P = 0.0077, respectively). The level of BAG2 in patients with PD was significantly correlated with Cross‐Culture Smell Identification Test score, which indicates olfactory dysfunction. We found that our biomarker model distinguishes PD with 87.5% diagnostic accuracy (area under the curve (AUC) = 0.875, P < 0.0001). Our result suggests BAG2 and cathepsin D as candidates for early‐diagnosis plasma biomarkers for PD. We provide the possibility of plasma biomarkers related to the autophagy pathway, by which decreased levels of BAG2 and cathepsin D might lead to dysfunction of autophagy.
Collapse
Affiliation(s)
- Juhee Kang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jae Whan Kim
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Hansol Heo
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jihyun Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, Korea
| | - Kwan Yong Park
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jung Han Yoon
- Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - Jaerak Chang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
69
|
Petrovic S, Arsic A, Ristic-Medic D, Cvetkovic Z, Vucic V. Lipid Peroxidation and Antioxidant Supplementation in Neurodegenerative Diseases: A Review of Human Studies. Antioxidants (Basel) 2020; 9:1128. [PMID: 33202952 PMCID: PMC7696060 DOI: 10.3390/antiox9111128] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 02/05/2023] Open
Abstract
Being characterized by progressive and severe damage in neuronal cells, neurodegenerative diseases (NDDs) are the major cause of disability and morbidity in the elderly, imposing a significant economic and social burden. As major components of the central nervous system, lipids play important roles in neural health and pathology. Disturbed lipid metabolism, particularly lipid peroxidation (LPO), is associated with the development of many NDDs, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), all of which show elevated levels of LPO products and LPO-modified proteins. Thus, the inhibition of neuronal oxidation might slow the progression and reduce the severity of NDD; natural antioxidants, such as polyphenols and antioxidant vitamins, seem to be the most promising agents. Here, we summarize current literature data that were derived from human studies on the effect of natural polyphenols and vitamins A, C, and E supplementation in patients with AD, PD, and ALS. Although these compounds may reduce the severity and slow the progression of NDD, research gaps remain in antioxidants supplementation in AD, PD, and ALS patients, which indicates that further human studies applying antioxidant supplementation in different forms of NDDs are urgently needed.
Collapse
Affiliation(s)
- Snjezana Petrovic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (A.A.); (D.R.-M.)
| | - Aleksandra Arsic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (A.A.); (D.R.-M.)
| | - Danijela Ristic-Medic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (A.A.); (D.R.-M.)
| | - Zorica Cvetkovic
- Department of Hematology, Clinical Hospital Center Zemun, 11000 Belgrade, Serbia;
- Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia
| | - Vesna Vucic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia; (S.P.); (A.A.); (D.R.-M.)
| |
Collapse
|
70
|
Magalingam KB, Radhakrishnan AK, Somanath SD, Md S, Haleagrahara N. Influence of serum concentration in retinoic acid and phorbol ester induced differentiation of SH-SY5Y human neuroblastoma cell line. Mol Biol Rep 2020; 47:8775-8788. [PMID: 33098048 DOI: 10.1007/s11033-020-05925-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/14/2020] [Indexed: 01/08/2023]
Abstract
Numerous protocols to establish dopaminergic phenotype in SH-SY5Y cells have been reported. In most of these protocols there are variations in concentration of serum used. In this paper, we compared the effects of high (10%), low (3%) and descending (2.5%/1%) serum concentration in differentiation medium containing different proportion of retinoic acid (RA) and 12-O-Tetradecanoylphorbol-13-acetate (TPA) or RA-only on the undifferentiated SH-SY5Y cells with regards to cell morphology, biochemical and gene expression alterations. Cells differentiated in culture medium containing low and descending serum concentrations showed increased number of neurite projections and reduced proliferation rates when compared to undifferentiated cells. The SH-SY5Y cells differentiated in culture medium containing 3% RA and low serum or descending (2.5%/1% RA/TPA) were found to be more susceptible to 6-hydroxydopamine (6-OHDA) induced cytotoxicity. Cells differentiated with RA/TPA or RA differentiated showed increased production of the α-synuclein (SNCA) neuroprotein and dopamine neurotransmitter compared to undifferentiated cells, regardless serum concentrations used. There was no significant difference in the expression of tyrosine hydroxylase (TH) gene between undifferentiated and differentiated SH-SY5Y cells. However, the expression of dopamine receptor D2 (DRD2) gene was markedly increased (p<0.05) in differentiated cells with 3% serum and RA only when compared to undifferentiated cells. In conclusion, to terminally differentiate SH-SY5Y cells to be used as a cell-based model to study Parkinson's disease (PD) to investigate molecular mechanisms and drug discovery, the optimal differentiation medium should contain 3% serum in RA-only.
Collapse
Affiliation(s)
| | - Ammu Kutty Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Malaysia, Bandar Sunway, Malaysia
- Pathology Division, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Sushela Devi Somanath
- Pathology Division, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Nagaraja Haleagrahara
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia.
- College of Public Health, Medicine and Veterinary Sciences, James Cook University, Townsville, Queensland, 4811, Australia.
| |
Collapse
|
71
|
Hall MFE, Church FC. Exercise for Older Adults Improves the Quality of Life in Parkinson's Disease and Potentially Enhances the Immune Response to COVID-19. Brain Sci 2020; 10:E612. [PMID: 32899958 PMCID: PMC7563553 DOI: 10.3390/brainsci10090612] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/26/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder brought about due to dopaminergic neuronal cell loss in the midbrain substantia nigra pars compacta region. PD presents most commonly in older adults and is a disorder of both motor and nonmotor dysfunction. The novel SARS-CoV-2 virus is responsible for the recent COVID-19 pandemic, and older individuals, those with preexisting medical conditions, or both have an increased risk of developing COVID-19 with more severe outcomes. People-with-Parkinson's (PwP) of advanced age can have both immune and autonomic nervous problems that potentially lead to pre-existing pulmonary dysfunction and higher infection risk, increasing the probability of contracting COVID-19. A lifestyle change involving moderate-intensity exercise has the potential to protect against SARS-CoV-2 through strengthening the immune system. In addition to a potential protective measure against SARS-CoV-2, exercise has been shown to improve quality-of-life (QoL) in PD patients. Recent studies provide evidence of exercise as both neuroprotective and neuroplastic. This article is a literature review investigating the role exercise plays in modifying the immune system, improving health outcomes in PwP, and potentially acting as a protective measure against SARS-Cov-2 infection. We conclude that exercise, when correctly performed, improves QoL and outcomes in PwP, and that the enhanced immune response from moderate-intensity exercise could potentially offer additional protection against COVID-19.
Collapse
Affiliation(s)
| | - Frank C. Church
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC 27599, USA;
| |
Collapse
|
72
|
Advani D, Gupta R, Tripathi R, Sharma S, Ambasta RK, Kumar P. Protective role of anticancer drugs in neurodegenerative disorders: A drug repurposing approach. Neurochem Int 2020; 140:104841. [PMID: 32853752 DOI: 10.1016/j.neuint.2020.104841] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/24/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
The disease heterogeneity and little therapeutic progress in neurodegenerative diseases justify the need for novel and effective drug discovery approaches. Drug repurposing is an emerging approach that reinvigorates the classical drug discovery method by divulging new therapeutic uses of existing drugs. The common biological background and inverse tuning between cancer and neurodegeneration give weight to the conceptualization of repurposing of anticancer drugs as novel therapeutics. Many studies are available in the literature, which highlights the success story of anticancer drugs as repurposed therapeutics. Among them, kinase inhibitors, developed for various oncology indications evinced notable neuroprotective effects in neurodegenerative diseases. In this review, we shed light on the salient role of multiple protein kinases in neurodegenerative disorders. We also proposed a feasible explanation of the action of kinase inhibitors in neurodegenerative disorders with more attention towards neurodegenerative disorders. The problem of neurotoxicity associated with some anticancer drugs is also highlighted. Our review encourages further research to better encode the hidden potential of anticancer drugs with the aim of developing prospective repurposed drugs with no toxicity for neurodegenerative disorders.
Collapse
Affiliation(s)
- Dia Advani
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rohan Gupta
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rahul Tripathi
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Sudhanshu Sharma
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Department of Biotechnology, Molecular Neuroscience and Functional Genomics Laboratory, Room# FW4TF3, Mechanical Engineering Building, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
73
|
Tang BL. RAB39B's role in membrane traffic, autophagy, and associated neuropathology. J Cell Physiol 2020; 236:1579-1592. [PMID: 32761840 DOI: 10.1002/jcp.29962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/19/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022]
Abstract
Neuropathological disorders are increasingly associated with dysfunctions in neuronal membrane traffic and autophagy, with defects among members of the Rab family of small GTPases implicated. Mutations in the human Xq28 localized gene RAB39B have been associated with X-linked neurodevelopmental defects including macrocephaly, intellectual disability, autism spectrum disorder (ASD), as well as rare cases of early-onset Parkinson's disease (PD). Despite the finding that RAB39B regulates GluA2 trafficking and could thus influence synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit composition, reasons for the wide-ranging neuropathological consequences associated with RAB39B defects have been unclear. Recent studies have now unraveled possible mechanisms underlying the neuropathological roles of this brain-enriched small GTPase. Studies in RAB39B knockout mice showed that RAB39B interacts with components of Class I phosphatidylinositol-3-kinase (PI3K) signaling. In its absence, the PI3K-AKT-mechanistic target of rapamycin signaling pathway in neural progenitor cells (NPCs) is hyperactivated, which promotes NPC proliferation, leading to macrocephaly and ASD. Pertaining to early-onset PD, a complex of C9orf72, Smith-Magenis syndrome chromosome region candidate 8 and WD repeat domain 41 that functions in autophagy has been identified as a guanine nucleotide exchange factor of RAB39B. Here, recent findings that have shed light on our mechanistic understanding of RAB39B's role in neurodevelopmental and neurodegenerative pathologies are reviewed. Caveats and unanswered questions are also discussed, and future perspectives outlined.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
74
|
Flynn JD, Gimmen MY, Dean DN, Lacy SM, Lee JC. Terminal Alkynes as Raman Probes of α-Synuclein in Solution and in Cells. Chembiochem 2020; 21:1582-1586. [PMID: 31960993 PMCID: PMC7269816 DOI: 10.1002/cbic.202000026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Indexed: 01/08/2023]
Abstract
Conformational changes in α-synuclein (α-syn) are central to its biological function and Parkinson's disease pathology. Here, terminal alkynes (homopropargylglycine) were employed as environmentally sensitive Raman probes at residues 1, 5, 116, and 127 to characterize soluble (disordered), micelle-bound (α-helical), and fibrillar (β-sheet) α-syn. Along with the full-length protein, a disease-related C-terminal truncation (1-115) was also studied. For the first time, β-sheet α-syn amyloid structure was detected by the amide-I band in N27 dopaminergic rat cells, where a reciprocal relationship between levels of fibrils and lipids was seen. Site-specific spectral features of the terminal alkynes also revealed the heterogeneity of the cellular environment. This work shows the versatility of Raman microspectroscopy and the power of unnatural amino acids in providing structural and residue-level insights in solution and in cells.
Collapse
Affiliation(s)
- Jessica D. Flynn
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892 (USA)
| | - Megan Y. Gimmen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892 (USA)
| | - Dexter N. Dean
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892 (USA)
| | - Shannon M. Lacy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892 (USA)
| | - Jennifer C. Lee
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892 (USA)
| |
Collapse
|
75
|
Leger C, Herbert M, DeSouza JFX. Non-motor Clinical and Biomarker Predictors Enable High Cross-Validated Accuracy Detection of Early PD but Lesser Cross-Validated Accuracy Detection of Scans Without Evidence of Dopaminergic Deficit. Front Neurol 2020; 11:364. [PMID: 32477243 PMCID: PMC7232850 DOI: 10.3389/fneur.2020.00364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/14/2020] [Indexed: 11/30/2022] Open
Abstract
Background: Early stage (preclinical) detection of Parkinson's disease (PD) remains challenged yet is crucial to both differentiate it from other disorders and facilitate timely administration of neuroprotective treatment as it becomes available. Objective: In a cross-validation paradigm, this work focused on two binary predictive probability analyses: classification of early PD vs. controls and classification of early PD vs. SWEDD (scans without evidence of dopamine deficit). It was hypothesized that five distinct model types using combined non-motor and biomarker features would distinguish early PD from controls with > 80% cross-validated (CV) accuracy, but that the diverse nature of the SWEDD category would reduce early PD vs. SWEDD CV classification accuracy and alter model-based feature selection. Methods: Cross-sectional, baseline data was acquired from the Parkinson's Progressive Markers Initiative (PPMI). Logistic regression, general additive (GAM), decision tree, random forest and XGBoost models were fitted using non-motor clinical and biomarker features. Randomized train and test data partitions were created. Model classification CV performance was compared using the area under the curve (AUC), sensitivity, specificity and the Kappa statistic. Results: All five models achieved >0.80 AUC CV accuracy to distinguish early PD from controls. The GAM (CV AUC 0.928, sensitivity 0.898, specificity 0.897) and XGBoost (CV AUC 0.923, sensitivity 0.875, specificity 0.897) models were the top classifiers. Performance across all models was consistently lower in the early PD/SWEDD analyses, where the highest performing models were XGBoost (CV AUC 0.863, sensitivity 0.905, specificity 0.748) and random forest (CV AUC 0.822, sensitivity 0.809, specificity 0.721). XGBoost detection of non-PD SWEDD matched 1-2 years curated diagnoses in 81.25% (13/16) cases. In both early PD/control and early PD/SWEDD analyses, and across all models, hyposmia was the single most important feature to classification; rapid eye movement behavior disorder (questionnaire) was the next most commonly high ranked feature. Alpha-synuclein was a feature of import to early PD/control but not early PD/SWEDD classification and the Epworth Sleepiness scale was antithetically important to the latter but not former. Interpretation: Non-motor clinical and biomarker variables enable high CV discrimination of early PD vs. controls but are less effective discriminating early PD from SWEDD.
Collapse
Affiliation(s)
- Charles Leger
- Department of Psychology, York University, Toronto, ON, Canada
- Neuroscience Diploma, York University, Toronto, ON, Canada
| | - Monique Herbert
- Department of Psychology, York University, Toronto, ON, Canada
| | - Joseph F. X. DeSouza
- Department of Psychology, York University, Toronto, ON, Canada
- Neuroscience Diploma, York University, Toronto, ON, Canada
- Centre for Vision Research, York University, Toronto, ON, Canada
- Department of Biology, York University, Toronto, ON, Canada
- Canadian Action and Perception Network (CAPnet), Vision: Science to Applications (VISTA), York University, Toronto, ON, Canada
| |
Collapse
|
76
|
Zhou H, Li S, Li C, Yang X, Li H, Zhong H, Lu JH, Lee SMY. Oxyphylla A Promotes Degradation of α-Synuclein for Neuroprotection via Activation of Immunoproteasome. Aging Dis 2020; 11:559-574. [PMID: 32489702 PMCID: PMC7220298 DOI: 10.14336/ad.2019.0612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/12/2019] [Indexed: 11/23/2022] Open
Abstract
Parkinson’s disease (PD), the second most common neurodegenerative disorder, is neuropathologically characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) and the presence of Lewy bodies in surviving neurons. α-synuclein (α-syn) is the major component of Lewy bodies and its deposition in neurons is critical pathological event in the pathogenesis of PD. Herein, we reported that Oxyphylla A, a novel lead compound from the fruit of Alpinia oxyphylla, significantly promoted α-syn degradation in a cellular PD model. When exploring the molecular pathways, we found that Oxyphylla A promoted α-syn degradation in a ubiquitin proteasome system (UPS)-dependent and autophagy-independent manner. We further confirmed that Oxyphylla A enhanced UPS activity by upregulating 20S subunit PSMB8 expression. A mechanism study revealed that Oxyphylla A activated the PKA/Akt/mTOR pathway to trigger PSMB8 expression and enhance UPS activity. Finally, we illustrated that Oxyphylla A alleviated the accumulation of both Triton-soluble and Triton-insoluble forms of α-syn and protected against α-syn-induced neurotoxicity in A53T α-syn transgenic mice. These findings suggest that the activation of UPS, via small molecular UPS enhancers including Oxyphylla A, may be a therapeutic strategy for intervention against PD and related diseases.
Collapse
Affiliation(s)
- Hefeng Zhou
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shengnan Li
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Chuwen Li
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xuanjun Yang
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China.,2Department of Biology, South University of Science and Technology, Shenzhen, China
| | - Haitao Li
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hanbing Zhong
- 2Department of Biology, South University of Science and Technology, Shenzhen, China
| | - Jia-Hong Lu
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Simon Ming-Yuen Lee
- 1State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
77
|
Molenaars M, Daniels EG, Meurs A, Janssens GE, Houtkooper RH. Mitochondrial cross-compartmental signalling to maintain proteostasis and longevity. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190414. [PMID: 32362258 DOI: 10.1098/rstb.2019.0414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lifespan in eukaryotic species can be prolonged by shifting from cellular states favouring growth to those favouring maintenance and stress resistance. For instance, perturbations in mitochondrial oxidative phosphorylation (OXPHOS) can shift cells into this latter state and extend lifespan. Because mitochondria rely on proteins synthesized from nuclear as well as mitochondrial DNA, they need to constantly send and receive messages from other compartments of the cell in order to function properly and maintain homeostasis, and lifespan extension is often dependent on this cross-compartmental signalling. Here, we describe the mechanisms of bi-directional mitochondrial cross-compartmental signalling resulting in proteostasis and longevity. These proteostasis mechanisms are highly context-dependent, governed by the origin and extent of stress. Furthermore, we discuss the translatability of these mechanisms and explore therapeutic developments, such as the antibiotic studies targeting mitochondria or mitochondria-derived peptides as therapies for age-related diseases such as neurodegeneration and cancer. This article is part of the theme issue 'Retrograde signalling from endosymbiotic organelles'.
Collapse
Affiliation(s)
- Marte Molenaars
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Eileen G Daniels
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Amber Meurs
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
78
|
Gómez-Benito M, Granado N, García-Sanz P, Michel A, Dumoulin M, Moratalla R. Modeling Parkinson's Disease With the Alpha-Synuclein Protein. Front Pharmacol 2020; 11:356. [PMID: 32390826 PMCID: PMC7191035 DOI: 10.3389/fphar.2020.00356] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein (α-Syn) is a key protein involved in Parkinson's disease (PD) pathology. PD is characterized by the loss of dopaminergic neuronal cells in the substantia nigra pars compacta and the abnormal accumulation and aggregation of α-Syn in the form of Lewy bodies and Lewy neurites. More precisely, the aggregation of α-Syn is associated with the dysfunctionality and degeneration of neurons in PD. Moreover, mutations in the SNCA gene, which encodes α-Syn, cause familial forms of PD and are the basis of sporadic PD risk. Given the role of the α-Syn protein in the pathology of PD, animal models that reflect the dopaminergic neuronal loss and the widespread and progressive formation of α-Syn aggregates in different areas of the brain constitute a valuable tool. Indeed, animal models of PD are important for understanding the molecular mechanisms of the disease and might contribute to the development and validation of new therapies. In the absence of animal models that faithfully reproduce human PD, in recent years, numerous animal models of PD based on α-Syn have been generated. In this review, we summarize the main features of the α-Syn pre-formed fibrils (PFFs) model and recombinant adeno-associated virus vector (rAAV) mediated α-Syn overexpression models, providing a detailed comparative analysis of both models. Here, we discuss how each model has contributed to our understanding of PD pathology and the advantages and weakness of each of them.
Collapse
Affiliation(s)
- Mónica Gómez-Benito
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Noelia Granado
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia García-Sanz
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Anne Michel
- UCB Biopharma, Neuroscience TA, Braine L'Alleud, Belgium
| | - Mireille Dumoulin
- Centre of Protein Engineering, InBios, University of Liege, Liège, Belgium
| | - Rosario Moratalla
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
79
|
Soto-Rojas LO, Martínez-Dávila IA, Luna-Herrera C, Gutierrez-Castillo ME, Lopez-Salas FE, Gatica-Garcia B, Soto-Rodriguez G, Bringas Tobon ME, Flores G, Padilla-Viveros A, Bañuelos C, Blanco-Alvarez VM, Dávila-Ayala J, Reyes-Corona D, Garcés-Ramírez L, Hidalgo-Alegria O, De La Cruz-lópez F, Martinez-Fong D. Unilateral intranigral administration of β-sitosterol β-D-glucoside triggers pathological α-synuclein spreading and bilateral nigrostriatal dopaminergic neurodegeneration in the rat. Acta Neuropathol Commun 2020; 8:56. [PMID: 32321590 PMCID: PMC7178762 DOI: 10.1186/s40478-020-00933-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/14/2020] [Indexed: 02/05/2023] Open
Abstract
The spreading and accumulation of α-synuclein and dopaminergic neurodegeneration, two hallmarks of Parkinson’s disease (PD), have been faithfully reproduced in rodent brains by chronic, oral administration of β-sitosterol β-D-glucoside (BSSG). We investigated whether a single injection of BSSG (6 μg BSSG/μL DMSO) in the left substantia nigra of Wistar rats causes the same effects. Mock DMSO injections and untreated rats formed control groups. We performed immunostainings against the pathological α-synuclein, the dopaminergic marker tyrosine hydroxylase (TH), the neuroskeleton marker β-III tubulin, the neurotensin receptor type 1 (NTSR1) as non-dopaminergic phenotype marker and Fluro-Jade C (F-J C) label for neurodegeneration. Using β-galactosidase (β-Gal) assay and active caspase-3 immunostaining, we assessed cell death mechanisms. Golgi-Cox staining was used to measure the density and types of dendritic spines of striatal medium spiny neurons. Motor and non-motor alterations were also evaluated. The study period comprised 15 to 120 days after the lesion. In the injured substantia nigra, BSSG caused a progressive α-synuclein aggregation and dopaminergic neurodegeneration caused by senescence and apoptosis. The α-synuclein immunoreactivity was also present within microglia cells. Decreased density of dopaminergic fibers and dendritic spines also occurred in the striatum. Remarkably, all the histopathological changes also appeared on the contralateral nigrostriatal system, and α-synuclein aggregates were present in other brain regions. Motor and non-motor behavioral alterations were progressive. Our data show that the stereotaxic BSSG administration reproduces PD α-synucleinopathy phenotype in the rat. This approach will aid in identifying the spread mechanism of α-synuclein pathology and validate anti-synucleinopathy therapies.
Collapse
|
80
|
Ultrasonography of the Vagus Nerve in the Diagnosis of Parkinson's Disease. PARKINSONS DISEASE 2020; 2020:2627471. [PMID: 32318257 PMCID: PMC7150709 DOI: 10.1155/2020/2627471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 11/29/2022]
Abstract
Background It is currently impossible to diagnose Parkinson's disease (PD) in the premotor phase even though at the time of motor symptom onset the number of already degenerated dopaminergic substantia nigra neurons is considerable. Degeneration of the dorsal nucleus of the vagus nerve (VN) has been reported early in the disease course, and it could lead to impaired function of the VN, resulting in certain nonmotor symptoms of PD. Therefore, we raised a hypothesis that the loss of VN neurons could result in a smaller diameter of the VN among PD patients. Methods 20 PD patients and 20 age- and gender-matched individuals without any neurodegenerative disease were enrolled in a pilot study. The diameters of the right and left VNs were measured using ultrasonography, their average was calculated, and the narrower VN diameter was noted separately. Results No difference was found between the PD and control groups neither in the average VN diameter (mean 1.17; 95% confidence interval (CI) 1.10–1.24 vs. 1.13; 1.07–1.18, mm; p=0.353) nor in the narrower VN diameter (mean 1.11; 95% confidence interval (CI) 1.02–1.20 vs. 1.07; 1.02–1.13, mm; p=0.421). The narrower VN diameter and the average VN diameter were not able to distinguish between PD patients and controls (area under curve (AUC) = 0.588, 95% CI = 0.408–0.767, and p=0.344; and AUC = 0.578, 95% CI = 0.396–0.759, and p=0.402). Conclusions To conclude, no differences were found in VN diameter between the PD and control groups. Therefore, our data do not support the hypothesis that PD could be associated with a smaller diameter of the VN.
Collapse
|
81
|
Kumari R, Kumar R, Kumar S, Singh AK, Hanpude P, Jangir D, Maiti TK. Amyloid aggregates of the deubiquitinase OTUB1 are neurotoxic, suggesting that they contribute to the development of Parkinson's disease. J Biol Chem 2020; 295:3466-3484. [PMID: 32005664 DOI: 10.1074/jbc.ra119.009546] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 01/18/2020] [Indexed: 01/10/2023] Open
Abstract
Parkinson's disease (PD) is a multifactorial malady and the second most common neurodegenerative disorder, characterized by loss of dopaminergic neurons in the midbrain. A hallmark of PD pathology is the formation of intracellular protein inclusions, termed Lewy bodies (LBs). Recent MS studies have shown that OTU deubiquitinase ubiquitin aldehyde-binding 1 (OTUB1), a deubiquitinating enzyme of the OTU family, is enriched together with α-synuclein in LBs from individuals with PD and is also present in amyloid plaques associated with Alzheimer's disease. In the present study, using mammalian cell cultures and a PD mouse model, along with CD spectroscopy, atomic force microscopy, immunofluorescence-based imaging, and various biochemical assays, we demonstrate that after heat-induced protein aggregation, OTUB1 reacts strongly with both anti-A11 and anti-osteocalcin antibodies, detecting oligomeric, prefibrillar structures or fibrillar species of amyloidogenic proteins, respectively. Further, recombinant OTUB1 exhibited high thioflavin-T and Congo red binding and increased β-sheet formation upon heat induction. The oligomeric OTUB1 aggregates were highly cytotoxic, characteristic of many amyloid proteins. OTUB1 formed inclusions in neuronal cells and co-localized with thioflavin S and with α-synuclein during rotenone-induced stress. It also co-localized with the disease-associated variant pS129-α-synuclein in rotenone-exposed mouse brains. Interestingly, OTUB1 aggregates were also associated with severe cytoskeleton damage, rapid internalization inside the neuronal cells, and mitochondrial damage, all of which contribute to neurotoxicity. In conclusion, the results of our study indicate that OTUB1 may contribute to LB pathology through its amyloidogenic properties.
Collapse
Affiliation(s)
- Raniki Kumari
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India; Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, Odisha 751024, India
| | - Roshan Kumar
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sanjay Kumar
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Abhishek Kumar Singh
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India
| | - Pranita Hanpude
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India
| | - Deepak Jangir
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India
| | - Tushar Kanti Maiti
- Functional Proteomics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, Faridabad 121001, India.
| |
Collapse
|
82
|
Negi N, Das BK. Decoding intrathecal immunoglobulins and B cells in the CNS: their synthesis, function, and regulation. Int Rev Immunol 2020; 39:67-79. [PMID: 31928379 DOI: 10.1080/08830185.2019.1711073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The discovery of an active lymphatic system in the meninges (dura mater) has opened up a wide range of possibilities for the role of CNS immunoglobulins in brain development in early fetal life or during infancy. The antibody-dependent and -independent functions of B cells in the immunopathogenesis of multiple sclerosis are not new to immunologists, yet their role in other neurodegenerative disorders such as Alzheimer's and Parkinson's disease is incompletely understood. Deep cervical lymph nodes have emerged as a candidate site for autosensitization against CNS antigens and have been shown to provide the right kind of milieu for the dynamic interaction of antigen-presenting cells, B cells, and T cells. The presence of different B cells in the lymph nodes and the production of natural autoantibodies by B-1 cells have definitely unlocked another piece of the puzzle. At a time when CD19 and CD20 monoclonal antibodies have shown remarkable results in ameliorating the relapse and progression of multiple sclerosis, it is imperative to dissect out the diversity in B cell populations inside the CNS to identify new targets to improve current treatment regimens for neurodegenerative diseases. This review highlights the origin, migration, function, and regulation of B cells and the production of intrathecal immunoglobulins considering the previous and current findings and taking into account the differences between a healthy state and the changes that occur during an inflammatory or autoimmune response.
Collapse
Affiliation(s)
- Neema Negi
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland, Galway, Ireland
| | - Bimal K Das
- HIV Immunology Section, Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
83
|
Longoni B, Fasciani I, Kolachalam S, Pietrantoni I, Marampon F, Petragnano F, Aloisi G, Coppolino MF, Rossi M, Scarselli M, Maggio R. Neurotoxic and Neuroprotective Role of Exosomes in Parkinson’s Disease. Curr Pharm Des 2020; 25:4510-4522. [DOI: 10.2174/1381612825666191113103537] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/13/2019] [Indexed: 01/06/2023]
Abstract
:Exosomes are extracellular vesicles produced by eukaryotic cells that are also found in most biological fluids and tissues. While they were initially thought to act as compartments for removal of cellular debris, they are now recognized as important tools for cell-to-cell communication and for the transfer of pathogens between the cells. They have attracted particular interest in neurodegenerative diseases for their potential role in transferring prion-like proteins between neurons, and in Parkinson’s disease (PD), they have been shown to spread oligomers of α-synuclein in the brain accelerating the progression of this pathology. A potential neuroprotective role of exosomes has also been equally proposed in PD as they could limit the toxicity of α-synuclein by clearing them out of the cells. Exosomes have also attracted considerable attention for use as drug vehicles. Being nonimmunogenic in nature, they provide an unprecedented opportunity to enhance the delivery of incorporated drugs to target cells. In this review, we discuss current knowledge about the potential neurotoxic and neuroprotective role of exosomes and their potential application as drug delivery systems in PD.
Collapse
Affiliation(s)
- Biancamaria Longoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Shivakumar Kolachalam
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Ilaria Pietrantoni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | | | - Francesco Petragnano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Gabriella Aloisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Maria F. Coppolino
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Mario Rossi
- Institute of Molecular Cell and Systems Biology, University of Glasgow, United Kingdom
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| |
Collapse
|
84
|
Tatullo M, Codispoti B, Spagnuolo G, Zavan B. Human Periapical Cyst-Derived Stem Cells Can Be A Smart "Lab-on-A-Cell" to Investigate Neurodegenerative Diseases and the Related Alteration of the Exosomes' Content. Brain Sci 2019; 9:E358. [PMID: 31817546 PMCID: PMC6955839 DOI: 10.3390/brainsci9120358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
Promising researches have demonstrated that the alteration of biological rhythms may be consistently linked to neurodegenerative pathologies. Parkinson's disease (PD) has a multifactorial pathogenesis, involving both genetic and environmental and/or molecular co-factors. Generally, heterogeneous alterations in circadian rhythm (CR) are a typical finding in degenerative processes, such as cell aging and death. Although numerous genetic phenotypes have been discovered in the most common forms of PD, it seems that severe deficiencies in synaptic transmission and high vesicular recycling are frequently found in PD patients. Neuron-to-neuron interactions are often ensured by exosomes, a specific type of extracellular vesicle (EV). Neuron-derived exosomes may carry several active compounds, including miRNAs: Several studies have found that circulating miRNAs are closely associated with an atypical oscillation of circadian rhythm genes, and they are also involved in the regulation of clock genes, in animal models. In this context, a careful analysis of neural-differentiated Mesenchymal Stem Cells (MSCs) and the molecular and genetic characterization of their exosome content, both in healthy cells and in PD-induced cells, could be a strategic field of investigation for early diagnosis and better treatment of PD and similar neurodegenerative pathologies. A novel MSC population, called human periapical cyst-mesenchymal stem cells (hPCy-MSCs), has demonstrated that it naively expresswa the main neuronal markers, and may differentiate towards functional neurons. Therefore, hPCy-MSCs can be considered of particular interest for testing of in vitro strategies to treat neurological diseases. On the other hand, the limitations of using stem cells is an issue that leads researchers to perform experimental studies on the exosomes released by MCSs. Human periapical cyst-derived mesenkymal stem cells can be a smart "lab-on-a-cell" to investigate neurodegenerative diseases and the related exosomes' content alteration.
Collapse
Affiliation(s)
- Marco Tatullo
- Marelli Health, Tecnologica Research Institute, Stem Cell Unit, 88900 Crotone, Italy;
- Department of Therapeutic Dentistry, Sechenov University Russia, 19c1 Moscow, Russia
| | - Bruna Codispoti
- Marelli Health, Tecnologica Research Institute, Stem Cell Unit, 88900 Crotone, Italy;
| | - Gianrico Spagnuolo
- Department of Therapeutic Dentistry, Sechenov University Russia, 19c1 Moscow, Russia
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples, 80138 Napoli, Italy
| | - Barbara Zavan
- Department of Medical Sciences, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| |
Collapse
|
85
|
Analysis of the Relationship between Type II Diabetes Mellitus and Parkinson's Disease: A Systematic Review. PARKINSONS DISEASE 2019; 2019:4951379. [PMID: 31871617 PMCID: PMC6906831 DOI: 10.1155/2019/4951379] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 12/31/2022]
Abstract
In the early sixties, a discussion started regarding the association between Parkinson's disease (PD) and type II diabetes mellitus (T2DM). Today, this potential relationship is still a matter of debate. This review aims to analyze both diseases concerning causal relationships and treatments. A total of 104 articles were found, and studies on animal and “in vitro” models showed that T2DM causes neurological alterations that may be associated with PD, such as deregulation of the dopaminergic system, a decrease in the expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), an increase in the expression of phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes 15 (PED/PEA-15), and neuroinflammation, as well as acceleration of the formation of alpha-synuclein amyloid fibrils. In addition, clinical studies described that Parkinson's symptoms were notably worse after the onset of T2DM, and seven deregulated genes were identified in the DNA of T2DM and PD patients. Regarding treatment, the action of antidiabetic drugs, especially incretin mimetic agents, seems to confer certain degree of neuroprotection to PD patients. In conclusion, the available evidence on the interaction between T2DM and PD justifies more robust clinical trials exploring this interaction especially the clinical management of patients with both conditions.
Collapse
|
86
|
Zhang S, Li H, Zheng L, Li H, Feng C, Zhang W. Identification of functional tRNA-derived fragments in senescence-accelerated mouse prone 8 brain. Aging (Albany NY) 2019; 11:10485-10498. [PMID: 31746776 PMCID: PMC6914438 DOI: 10.18632/aging.102471] [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: 07/27/2019] [Accepted: 11/08/2019] [Indexed: 01/06/2023]
Abstract
Transfer RNA-derived fragments (tRFs) are known to contribute to multiple illnesses, including cancers, viral infections, and age-related neurodegeneration. In this study, we used senescence-accelerated mouse prone 8 (SAMP8) as a model of neurodegenerative disorders such as Alzheimer’s disease and Parkinson’s disease, and a control, the senescence-accelerated mouse resistant 1 (SAMR1) model, to comprehensively explore differences in tRF expression between them. We discovered 570 tRF transcripts among which eight were differentially expressed. We then obtained 110 potential target genes in a miRNA-like pattern. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation suggest that these target genes participate in a variety of brain functions; e.g., synapse formation (GO: 0045202) and the synaptic vesicle cycle pathway. We further assessed in detail those tRFs whose miRNA-like pattern was most likely to promote the progression of either Alzheimer’s or Parkinson’s disease, such as AS-tDR-011775 acting on Mobp and Park2. Our findings suggest the eight dysregulated tRFs we uncovered here may be beneficially exploited as potential diagnostic biomarkers and/or therapeutic targets to treat age-related brain diseases.
Collapse
Affiliation(s)
- Shuai Zhang
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Beijing Normal University, Zhuhai, Guangdong 519087, China.,Engineering Research Center of Natural Medicine, Ministry of Education, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China
| | - Hejian Li
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Beijing Normal University, Zhuhai, Guangdong 519087, China.,Engineering Research Center of Natural Medicine, Ministry of Education, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China
| | - Ling Zheng
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Beijing Normal University, Zhuhai, Guangdong 519087, China.,Engineering Research Center of Natural Medicine, Ministry of Education, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China
| | - Hong Li
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Beijing Normal University, Zhuhai, Guangdong 519087, China.,Engineering Research Center of Natural Medicine, Ministry of Education, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China
| | - Chengqiang Feng
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Beijing Normal University, Zhuhai, Guangdong 519087, China.,Engineering Research Center of Natural Medicine, Ministry of Education, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China
| | - Wensheng Zhang
- Zhuhai Branch of State Key Laboratory of Earth Surface Processes and Resource Ecology, Beijing Normal University, Zhuhai, Guangdong 519087, China.,Engineering Research Center of Natural Medicine, Ministry of Education, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,Beijing Key Laboratory of Traditional Chinese Medicine Protection and Utilization, Faculty of Geographical Science, Beijing Normal University, Beijing 100875, China.,National and Local United Engineering Research Center for Panax Notoginseng Resources Protection and Utilization Technology, Kunming, Yunnan 650000, China
| |
Collapse
|
87
|
Mallah K, Quanico J, Raffo-Romero A, Cardon T, Aboulouard S, Devos D, Kobeissy F, Zibara K, Salzet M, Fournier I. Mapping Spatiotemporal Microproteomics Landscape in Experimental Model of Traumatic Brain Injury Unveils a link to Parkinson's Disease. Mol Cell Proteomics 2019; 18:1669-1682. [PMID: 31204315 PMCID: PMC6683007 DOI: 10.1074/mcp.ra119.001604] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major health concerns with no clinically-approved FDA drug available for therapeutic intervention. Several genomics and neuroproteomics studies have been employed to decipher the underlying pathological mechanisms involved that can serve as potential neurotherapeutic targets and unveil a possible underlying relation of TBI to other secondary neurological disorders. In this work, we present a novel high throughput systems biology approach using a spatially resolved microproteomics platform conducted on different brain regions in an experimental rat model of moderate of controlled cortical injury (CCI) at a temporal pattern postinjury (1 day, 3 days, 7 days, and 10 days). Mapping the spatiotemporal landscape of signature markers in TBI revealed an overexpression of major protein families known to be implicated in Parkinson's disease (PD) such as GPR158, HGMB1, synaptotagmin and glutamate decarboxylase in the ipsilateral substantia nigra. In silico bioinformatics docking experiments indicated the potential correlation between TBI and PD through alpha-synuclein. In an in vitro model, stimulation with palmitoylcarnitine triggered an inflammatory response in macrophages and a regeneration processes in astrocytes which also further confirmed the in vivo TBI proteomics data. Taken together, this is the first study to assess the microproteomics landscape in TBI, mainly in the substantia nigra, thus revealing a potential predisposition for PD or Parkinsonism post-TBI.
Collapse
Affiliation(s)
- Khalil Mallah
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France; §ER045, PRASE, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Jusal Quanico
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Antonella Raffo-Romero
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Tristan Cardon
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - Soulaimane Aboulouard
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France
| | - David Devos
- ¶Department of Neurology, Expert center for Parkinson's disease, Department of Pharmacology, University of Lille, CHU LILLE, INSERM UMR_S 1171, LICEND, France
| | - Firas Kobeissy
- ‖Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kazem Zibara
- §ER045, PRASE, Laboratory of Stem Cells, Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Michel Salzet
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France.
| | - Isabelle Fournier
- ‡Université de Lille, INSERM, U1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse (PRISM), F-59000 Lille, France.
| |
Collapse
|
88
|
Gupta AK, Pokhriyal R, Das U, Khan MI, Ratna Kumar D, Gupta R, Chadda RK, Ramachandran R, Goyal V, Tripathi M, Hariprasad G. Evaluation of α-synuclein and apolipoprotein E as potential biomarkers in cerebrospinal fluid to monitor pharmacotherapeutic efficacy in dopamine dictated disease states of Parkinson's disease and schizophrenia. Neuropsychiatr Dis Treat 2019; 15:2073-2085. [PMID: 31410011 PMCID: PMC6650621 DOI: 10.2147/ndt.s205550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/05/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Dopamine plays an important role in the disease pathology of Parkinson's disease and schizophrenia. These two neuropsychiatric disorders represent disease end points of the dopaminergic spectrum where Parkinson's disease represents dopamine deficit and schizophrenia represents dopamine hyperactivity in the mid-brain. Therefore, current treatment strategies aim to restore normal dopamine levels. However, during treatment patients develop adverse effects due to overshooting of physiological levels of dopamine leading to psychosis in Parkinson's disease, and extrapyramidal symptoms in schizophrenia. Absence of any laboratory tests hampers modulation of pharmacotherapy. Apolipoprotein E and α-synuclein have an important role in the neuropathology of these two diseases. The objective of this study was to evaluate cerebrospinal fluid (CSF) concentrations of apolipoprotein E and α-synuclein in patients with these two diseases so that they may serve as biomarkers to monitor therapy in Parkinson's disease and schizophrenia. METHODS Drug-naïve Parkinson's disease patients and Parkinson's disease patients treated with dopaminergic therapy, neurological controls, schizophrenic patients treated with antidopaminergic therapy, and drug-naïve schizophrenic patients were recruited for the study and CSF was collected. Enzyme-linked immunosorbent assays were carried out to estimate the concentrations of apolipoprotein E and α-synuclein. Pathway analysis was done to establish a possible role of these two proteins in various pathways in these two dopamine dictated diseases. RESULTS Apolipoprotein E and α-synuclein CSF concentrations have an inverse correlation along the entire dopaminergic clinical spectrum. Pathway analysis convincingly establishes a plausible hypothesis for their co-regulation in the pathogenesis of Parkinson's disease and schizophrenia. Each protein by itself or as a combination has encouraging sensitivity and specificity values of more than 55%. CONCLUSION The dynamic variation of these two proteins along the spectrum is ideal for them to be pursued as pharmacotherapeutic biomarkers in CSF to monitor pharmacological efficacy in Parkinson's disease and schizophrenia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Vinay Goyal
- Department of Neurology, All India Institute of Medical Sciences, New Delhi110029, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi110029, India
| | | |
Collapse
|
89
|
Kelley CM, Ginsberg SD, Alldred MJ, Strupp BJ, Mufson EJ. Maternal Choline Supplementation Alters Basal Forebrain Cholinergic Neuron Gene Expression in the Ts65Dn Mouse Model of Down Syndrome. Dev Neurobiol 2019; 79:664-683. [PMID: 31120189 PMCID: PMC6756931 DOI: 10.1002/dneu.22700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/12/2022]
Abstract
Down syndrome (DS), trisomy 21, is marked by intellectual disability and a premature aging profile including degeneration of the basal forebrain cholinergic neuron (BFCN) projection system, similar to Alzheimer's disease (AD). Although data indicate that perinatal maternal choline supplementation (MCS) alters the structure and function of these neurons in the Ts65Dn mouse model of DS and AD (Ts), whether MCS affects the molecular profile of vulnerable BFCNs remains unknown. We investigated the genetic signature of BFCNs obtained from Ts and disomic (2N) offspring of Ts65Dn dams maintained on a MCS diet (Ts+, 2N+) or a choline normal diet (ND) from mating until weaning, then maintained on ND until 4.4-7.5 months of age. Brains were then collected and prepared for choline acetyltransferase (ChAT) immunohistochemistry and laser capture microdissection followed by RNA extraction and custom-designed microarray analysis. Findings revealed upregulation of select transcripts in classes of genes related to the cytoskeleton (Tubb4b), AD (Cav1), cell death (Bcl2), presynaptic (Syngr1), immediate early (Fosb, Arc), G protein signaling (Gabarap, Rgs10), and cholinergic neurotransmission (Chrnb3) in Ts compared to 2N mice, which were normalized with MCS. Moreover, significant downregulation was seen in select transcripts associated with the cytoskeleton (Dync1h1), intracellular signaling (Itpka, Gng3, and Mlst8), and cell death (Ccng1) in Ts compared to 2N mice that was normalized with MCS. This study provides insight into genotype-dependent differences and the effects of MCS at the molecular level within a key vulnerable cell type in DS and AD.
Collapse
Affiliation(s)
- Christy M. Kelley
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA
- Department of Neuroscience & Physiology, NYU Langone School of Medicine, New York, NY, USA
- NYU Neuroscience Institute, NYU Langone School of Medicine, New York, NY, USA
| | - Melissa J. Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, NYU Langone School of Medicine, New York, NY, USA
| | - Barbara J. Strupp
- Division of Nutritional Sciences and Department of Psychology, Cornell University, Ithaca, NY, USA
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
90
|
Martínez-Orozco H, Mariño L, Uceda AB, Ortega-Castro J, Vilanova B, Frau J, Adrover M. Nitration and Glycation Diminish the α-Synuclein Role in the Formation and Scavenging of Cu 2+-Catalyzed Reactive Oxygen Species. ACS Chem Neurosci 2019; 10:2919-2930. [PMID: 30973706 DOI: 10.1021/acschemneuro.9b00142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Human α-synuclein is a small monomeric protein (140 residues) essential to maintain the function of the dopaminergic neurons and the neuronal redox balance. However, it holds a dark side since it is able to clump inside the neurons forming insoluble aggregates known as Lewy bodies, which are considered the hallmark of Parkinson's disease. Sporadic mutations and nonenzymatic post-translational modifications are well-known to stimulate the formation of Lewy bodies. Yet, the effect of nonenzymatic post-translational modifications on the function of α-synuclein has been studied less intense. Therefore, here we study how nitration and glycation mediated by methylglyoxal affect the redox features of α-synuclein. Both diminish the ability of α-synuclein to chelate Cu2+, except when Nε-(carboxyethyl)lysine or Nε-(carboxymethyl)lysine (two advanced glycation end products highly prevalent in vivo) are formed. This results in a lower capacity to prevent the Cu-catalyzed ascorbic acid degradation and to delay the formation of H2O2. However, only methylglyoxal was able to abolish the ability of α-synuclein to inhibit the free radical release. Both nitration and glycation enhanced the α-synuclein availability to be damaged by O2•-, although glycation made α-synuclein less reactive toward HO•. Our data represent the first report describing how nonenzymatic post-translational modifications might affect the redox function of α-synuclein, thus contributing to a better understanding of its pathological implications.
Collapse
Affiliation(s)
- Humberto Martínez-Orozco
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Laura Mariño
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Ana Belén Uceda
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Joaquín Ortega-Castro
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Bartolomé Vilanova
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Juan Frau
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Miquel Adrover
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| |
Collapse
|
91
|
Honarmand S, Dabirmanesh B, Amanlou M, Khajeh K. The interaction of several herbal extracts with α-synuclein: Fibril formation and surface plasmon resonance analysis. PLoS One 2019; 14:e0217801. [PMID: 31185031 PMCID: PMC6559707 DOI: 10.1371/journal.pone.0217801] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/17/2019] [Indexed: 11/18/2022] Open
Abstract
Proteins from their native conformation convert into highly ordered fibrillar aggregation under particular conditions; that are described as amyloid fibrils. α-Synuclein (α-Syn) is a small natively unfolded protein that its fibrillation is the causative factor of Parkinson's disease. One important approach in the development of therapeutic agents is the use of small molecules (such as flavonoids) that could specifically and efficiently inhibit the aggregation process. In this study the effect of few herbal extract (Berberis, Quercus robur, Zizyphus vulgaris, Salix aegyptica) containing flavonoids were investigated on fibril formation of α-syn by using conventional methods such as ThT fluorescence, circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM). The interaction of extracts were also analysed by surface plasmon resonance (SPR). Among extracts, Salix aegyptica revealed the highest inhibitory effect on fibril formation. As expected, Salix aegyptica extract also exhibited the highest affinity toward α-syn. Cell viability using MTT assay revealed that fibrils alone were more toxic than those containing the extract. Overall, we demonstrated that the affinity of compounds used in this study corresponds to their ability to arrest fibrillation and reduce cellular toxicity of α-syn fibrils.
Collapse
Affiliation(s)
- Shokouh Honarmand
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University,Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University,Tehran, Iran
- * E-mail:
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University,Tehran, Iran
| |
Collapse
|
92
|
Dal Ben M, Bongiovanni R, Tuniz S, Fioriti E, Tiribelli C, Moretti R, Gazzin S. Earliest Mechanisms of Dopaminergic Neurons Sufferance in a Novel Slow Progressing Ex Vivo Model of Parkinson Disease in Rat Organotypic Cultures of Substantia Nigra. Int J Mol Sci 2019; 20:2224. [PMID: 31064126 PMCID: PMC6539377 DOI: 10.3390/ijms20092224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022] Open
Abstract
The current treatments of Parkinson disease (PD) are ineffective mainly due to the poor understanding of the early events causing the decline of dopaminergic neurons (DOPAn). To overcome this problem, slow progressively degenerating models of PD allowing the study of the pre-clinical phase are crucial. We recreated in a short ex vivo time scale (96 h) all the features of human PD (needing dozens of years) by challenging organotypic culture of rat substantia nigra with low doses of rotenone. Thus, taking advantage of the existent knowledge, the model was used to perform a time-dependent comparative study of the principal possible causative molecular mechanisms undergoing DOPAn demise. Alteration in the redox state and inflammation started at 3 h, preceding the reduction in DOPAn number (pre-diagnosis phase). The number of DOPAn declined to levels compatible with diagnosis only at 12 h. The decline was accompanied by a persistent inflammation and redox imbalance. Significant microglia activation, apoptosis, a reduction in dopamine vesicle transporters, and the ubiquitination of misfolded protein clearance pathways were late (96 h, consequential) events. The work suggests inflammation and redox imbalance as simultaneous early mechanisms undergoing DOPAn sufferance, to be targeted for a causative treatment aimed to stop/delay PD.
Collapse
Affiliation(s)
- Matteo Dal Ben
- Department of Medical, Surgical, and Health Sciences, University of Trieste, 34100 Trieste, Italy.
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | | | - Simone Tuniz
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | - Emanuela Fioriti
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | - Claudio Tiribelli
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| | - Rita Moretti
- Neurology Clinic, Department of Medical, Surgical, and Health Sciences, University of Trieste, 34100 Trieste, Italy.
| | - Silvia Gazzin
- Fondazione Italiana Fegato, AREA Science Park, 34149 Trieste, Italy.
| |
Collapse
|
93
|
Kharrazian D, Herbert M, Vojdani A. The Associations between Immunological Reactivity to the Haptenation of Unconjugated Bisphenol A to Albumin and Protein Disulfide Isomerase with Alpha-Synuclein Antibodies. TOXICS 2019; 7:toxics7020026. [PMID: 31064082 PMCID: PMC6630361 DOI: 10.3390/toxics7020026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
Patients with Parkinson’s disease (PD) have increased susceptibility to bisphenol A (BPA) exposure since they have an impaired biotransformation capacity to metabolize BPA. PD subjects have reduced levels of conjugated BPA compared to controls. Reduced ability to conjugate BPA provides increased opportunity for unconjugated BPA to bind to albumin in human serum and protein disulfide isomerase on neurons. Once unconjugated BPA binds to proteins, it changes the allosteric structure of the newly configured protein leading to protein misfolding and the ability of the newly configured protein to act as a neoantigen. Once this neoantigen is formed, the immune system produces antibodies against it. The goal of our research was to investigate associations between unconjugated BPA bound to human serum albumin (BPA–HSA) antibodies and alpha-synuclein antibodies and between Protein Disulfide Isomerase (PDI) antibodies and alpha-synuclein antibodies. Enzyme–linked immunosorbent assay was used to determine the occurrences of alpha-synuclein antibodies, antibodies to BPA–HSA adducts, and PDI antibodies in the sera of blood donors. Subjects that exhibited high levels of unconjugated BPA–HSA antibodies or PDI antibodies had correlations and substantial risk for also exhibiting high levels of alpha-synuclein antibodies (p < 0.0001). We conclude that there are significant associations and risks between antibodies to BPA–HSA adducts and PDI antibodies for developing alpha-synuclein antibodies.
Collapse
Affiliation(s)
- Datis Kharrazian
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
- TRANSCEND Research Laboratory, Department of Neurology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA.
- Department of Preventive Medicine, Loma Linda University School of Medicine, 24785 Stewart Street, Loma Linda, CA 92354, USA.
| | - Martha Herbert
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
- TRANSCEND Research Laboratory, Department of Neurology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA.
| | - Aristo Vojdani
- Department of Preventive Medicine, Loma Linda University School of Medicine, 24785 Stewart Street, Loma Linda, CA 92354, USA.
- Immunosciences Lab., Inc., 822 S. Robertson Boulevard, Suite 312, Los Angeles, CA 90035, USA.
| |
Collapse
|
94
|
O'Leary EI, Lee JC. Interplay between α-synuclein amyloid formation and membrane structure. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2019; 1867:483-491. [PMID: 30287222 PMCID: PMC6445794 DOI: 10.1016/j.bbapap.2018.09.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 12/23/2022]
Abstract
Amyloid formation is a pathological hallmark of many neurodegenerative diseases, including Alzheimer's, Parkinson's, and Huntington's. While it is unknown how these disorders are initiated, in vitro and cellular experiments confirm the importance of membranes. Ubiquitous in vivo, membranes induce conformational changes in amyloidogenic proteins and in some cases, facilitate aggregation. Reciprocally, perturbations in the bilayer structure can be induced by amyloid formation. Here, we review studies in the last 10 years describing α-synuclein (α-syn) and its interactions with membranes, detailing the roles of anionic and zwitterionic lipids in aggregation, and their contribution to Parkinson's disease. We summarize the impact of α-syn - comparing monomeric, oligomeric, and fibrillar forms - on membrane structure, and the effect of membrane remodeling on amyloid formation. Finally, perspective on future studies investigating the interplay between α-syn aggregation and membranes is discussed. This article is part of a Special Issue entitled: Amyloids.
Collapse
Affiliation(s)
- Emma I O'Leary
- Laboratory of Protein Conformation and Dynamics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jennifer C Lee
- Laboratory of Protein Conformation and Dynamics, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
95
|
Masutani K, Yamamori Y, Kim K, Matubayasi N. Free-energy analysis of the hydration and cosolvent effects on the β-sheet aggregation through all-atom molecular dynamics simulation. J Chem Phys 2019; 150:145101. [DOI: 10.1063/1.5088395] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Keiichi Masutani
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Yu Yamamori
- Artificial Intelligence Research Center and Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Koto, Tokyo 135-0064, Japan
| | - Kang Kim
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Nobuyuki Matubayasi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
- Elements Strategy Initiative for Catalysts and Batteries, Kyoto University, Katsura, Kyoto 615-8520, Japan
| |
Collapse
|
96
|
Abeyawardhane DL, Heitger DR, Fernández RD, Forney AK, Lucas HR. C-Terminal Cu II Coordination to α-Synuclein Enhances Aggregation. ACS Chem Neurosci 2019; 10:1402-1410. [PMID: 30384594 DOI: 10.1021/acschemneuro.8b00448] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The structurally dynamic amyloidogenic protein α-synuclein (αS) is universally recognized as a key player in Parkinson's disease (PD). Copper, which acts as a neuronal signaling agent, is also an effector of αS structure, aggregation, and localization in vivo. In humans, αS is known to carry an acetyl group on the starting methionine residue, capping the N-terminal free amine which was a known high-affinity CuII binding site. We now report the first detailed characterization data using electron paramagnetic resonance (EPR) spectroscopy to describe the CuII coordination modes of N-terminally acetylated αS (NAcαS). Through use of EPR hyperfine structure analyses and the Peisach-Blumberg correlation, an N3O1 binding mode was established that involves the single histidine residue at position 50 and a lower population of a second CuII-binding mode that may involve a C-terminal contribution. We additionally generated an N-terminally acetylated disease-relevant variant, NAcH50Q, that promotes a shift in the CuII binding site to the C-terminus of the protein. Moreover, fibrillar NAcH50Q-CuII exhibits enhanced parallel β-sheet character and increased hydrophobic surface area compared to NAcαS-CuII and to both protein variants that lack a coordinated cupric ion. The results presented herein demonstrate the differential impact of distinct CuII binding sites within NAcαS, revealing that C-terminal CuII binding exacerbates the structural consequences of the H50Q missense mutation. Likewise, the global structural modifications that result from N-terminal capping augment the properties of CuII coordination. Hence, consideration of the effect of CuII on NAcαS and NAcH50Q misfolding may shed light on the extrinsic or environmental factors that influence PD pathology.
Collapse
Affiliation(s)
| | - Denver R. Heitger
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Ricardo D. Fernández
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Ashley K. Forney
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Heather R. Lucas
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
97
|
Fuzzati-Armentero MT, Cerri S, Blandini F. Peripheral-Central Neuroimmune Crosstalk in Parkinson's Disease: What Do Patients and Animal Models Tell Us? Front Neurol 2019; 10:232. [PMID: 30941089 PMCID: PMC6433876 DOI: 10.3389/fneur.2019.00232] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
The brain is no longer considered an immune privileged organ and neuroinflammation has long been associated with Parkinson's disease. Accumulating evidence demonstrates that innate and adaptive responses take place in the CNS. The extent to which peripheral immune alterations impacts on the CNS, or vice and versa, is, however, still a matter of debate. Gaining a better knowledge of the molecular and cellular immune dysfunctions present in these two compartments and clarifying their mutual interactions is a fundamental step in understanding and preventing Parkinson's disease (PD) pathogenesis. This review provides an overview of the current knowledge on inflammatory processes evidenced both in PD patients and in toxin-induced animal models of the disease. It discusses differences and similarities between human and animal studies in the context of neuroinflammation and immune responses and how they have guided therapeutic strategies to slow down disease progression. Future longitudinal studies are necessary and can help gain a better understanding on peripheral-central nervous system crosstalk to improve therapeutic strategies for PD.
Collapse
|
98
|
Jo MG, Ikram M, Jo MH, Yoo L, Chung KC, Nah SY, Hwang H, Rhim H, Kim MO. Gintonin Mitigates MPTP-Induced Loss of Nigrostriatal Dopaminergic Neurons and Accumulation of α-Synuclein via the Nrf2/HO-1 Pathway. Mol Neurobiol 2019; 56:39-55. [PMID: 29675576 DOI: 10.1007/s12035-018-1020-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/16/2018] [Indexed: 01/08/2023]
Abstract
Gintonin, a ginseng-derived glycolipoprotein isolated from ginseng, has been shown to be neuroprotective in several neurological disorders such as Alzheimer's disease models and depressive-like behaviors. In this study, we sought to investigate the potential protective mechanisms of gintonin in an in vivo MPTP and in vitro MPP+-mediated Parkinson's disease (PD) model. We hypothesized that activation of nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1, potential therapeutic targets for neurodegeneration) with gintonin could abrogate PD-associated neurotoxicity by modulating the accumulation of α-synuclein, neuroinflammation, and apoptotic cell death in an MPTP/MPP+ models of PD. Our in vivo and in vitro findings suggest that the neuroprotective effects of gintonin were associated with the regulation of the Nrf2/HO-1 pathway, which regulated the expression of proinflammatory cytokines and nitric oxide synthase and apoptotic markers in the substantia nigra and striatum of the mice. Moreover, the neuroprotective effects of gintonin were also associated with a reduction in α-synuclein accumulation in the mouse substantia nigra and striatum. The neuroprotective effects of gintonin were further validated by analyzing the effects of gintonin on MPP+-treated SH-SY5Y cells, which confirmed the protective effects of gintonin. It remains for future basic and clinical research to determine the potential use of gintonin in Parkinson's disease. However, to the best of our knowledge, marked alterations in biochemical and morphological setup of midbrain dopaminergic pathways by gintonin in MPTP mice model have not been previously reported. We believe that gintonin might be explored as an important therapeutic agent in the treatment of PD.
Collapse
Affiliation(s)
- Min Gi Jo
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju, 52802, Republic of Korea
| | - Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju, 52802, Republic of Korea
| | - Myeung Hoon Jo
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju, 52802, Republic of Korea
| | - Lang Yoo
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hongik Hwang
- Center for Neuroscience, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju, 52802, Republic of Korea.
- Division of Life Science and Applied Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
99
|
Matubayasi N, Masutani K. Energetics of cosolvent effect on peptide aggregation. Biophys Physicobiol 2019; 16:185-195. [PMID: 31984171 PMCID: PMC6975910 DOI: 10.2142/biophysico.16.0_185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/16/2019] [Indexed: 12/01/2022] Open
Abstract
The cosolvent effect on the equilibrium of peptide aggregation is reviewed from the energetic perspective. It is shown that the excess chemical potential is stationary against the variation of the distribution function for the configuration of a flexible solute species and that the derivative of the excess chemical potential with respect to the cosolvent concentration is determined by the corresponding derivative of the solvation free energy averaged over the solute configurations. The effect of a cosolvent at low concentrations on a chemical equilibrium can then be addressed in terms of the difference in the solvation free energy between pure-water solvent and the mixed solvent with the cosolvent, and illustrative analyses with all-atom model are presented for the aggregation of an 11-residue peptide by employing the energy-representation method to compute the solvation free energy. The solvation becomes more favorable with addition of the urea or DMSO cosolvent, and the extent of stabilization is smaller for larger aggregate. This implies that these cosolvents inhibit the formation of an aggregate, and the roles of such interaction components as the electrostatic, van der Waals, and excluded-volume are discussed.
Collapse
Affiliation(s)
- Nobuyuki Matubayasi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University
| | - Keiichi Masutani
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University
| |
Collapse
|
100
|
When safeguarding goes wrong: Impact of oxidative stress on protein homeostasis in health and neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 114:221-264. [PMID: 30635082 DOI: 10.1016/bs.apcsb.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular redox status is an established player in many different cellular functions. The buildup of oxidants within the cell is tightly regulated to maintain a balance between the positive and negative outcomes of cellular oxidants. Proteins are highly sensitive to oxidation, since modification can cause widespread unfolding and the formation of toxic aggregates. In response, cells have developed highly regulated systems that contribute to the maintenance of both the global redox status and protein homeostasis at large. Changes to these systems have been found to correlate with aging and age-related disorders, such as neurodegenerative pathologies. This raises intriguing questions as to the source of the imbalance in the redox and protein homeostasis systems, their interconnectivity, and their role in disease progression. Here we focus on the crosstalk between the redox and protein homeostasis systems in neurodegenerative diseases, specifically in Alzheimer's, Parkinson's, and ALS. We elaborate on some of the main players of the stress response systems, including the master regulators of oxidative stress and the heat shock response, Nrf2 and Hsf1, which are essential features of protein folding, and mediators of protein turnover. We illustrate the elegant mechanisms used by these components to provide an immediate response, including protein plasticity controlled by redox-sensing cysteines and the recruitment of naive proteins to the redox homeostasis array that act as chaperons in an ATP-independent manner.
Collapse
|