1
|
Liu R, Wu Q, Wu C, Qu Y, Fang Y, De J, Fan R, Song W. Metabolic signatures of metabolites of the purine degradation pathway in human plasma using HILIC UHPLC-HRMS. J Pharm Biomed Anal 2024; 251:116451. [PMID: 39217702 DOI: 10.1016/j.jpba.2024.116451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/16/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The metabolic disorders in the purine degradation pathway have proven to be closely associated with several human diseases. However, the etiology is not yet fully understood. Profile assay of purine intermediates and uric acid involved in the metabolic pathway can provide additional insight into the nature and severity of related diseases. Purine metabolites are endogenous chemicals with high hydrophilicity, polarity, and similar structures, thus there is a great need for a specific method to quantify them directly in biological fluids with a short running time. Herein, eight purine degradation pathway metabolites, including xanthine, hypoxanthine, guanine, xanthosine, inosine, guanosine, adenosine and uric acid, in human plasma were quantitatively measured using hydrophilic interaction chromatography-tandem high-resolution mass spectrometry (HILIC-HRMS) in a short running time of 10 min. The method was systematically validated for specificity, linearity of the calibration curve, the limit of detection, the limit of quantification, the lower limit of quantification, precision, accuracy, extraction recovery, matrix effect, and stability. The results showed that the method was linear (R2 > 0.99), accurate (the intra- and inter-day recoveries of all analytes ranged from 90.0 % to 110.0 %), and precise (the intra- and inter-day precisions were less than 6.7 % and 8.9 %, respectively) with the lower limits of quantification ranging from 3 to 10,000 ng/mL. The extraction recoveries and matrix effects were repeatable and stable. All the analytes were stable in the autosampler and could be subject to three freeze-thaw cycles. The developed method was ultimately applied to 100 plasma specimens from healthy individuals. The results showed that the concentrations of different purine metabolites varied dramatically in plasma specimens. Diet and body mass index (BMI) were the most significant factors determining purine levels, followed by drinking and sex. Age, smoking and bedtime showed a very weak correlation with purine metabolism. The findings of the present work reveal the characteristics of purine metabolism in human plasma under non-pathological conditions. The results also highlight the factors that can cause changes in purine metabolism, which are useful in developing effective treatment strategies for metabolic disorders of purines, particularly for those caused by lifestyle factors.
Collapse
Affiliation(s)
- Rui Liu
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Qingke Wu
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Chuanlong Wu
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Yingnan Qu
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Yanming Fang
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Jiyangzong De
- School of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Ronghua Fan
- School of Public Health, Shenyang Medical College, Shenyang 110034, China; Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang 110034, China.
| | - Wenjing Song
- School of Public Health, Shenyang Medical College, Shenyang 110034, China.
| |
Collapse
|
2
|
He K, Zhao Z, Zhang J, Li D, Wang S, Liu Q. Cholesterol Metabolism in Neurodegenerative Diseases. Antioxid Redox Signal 2024; 41:1051-1072. [PMID: 38842175 DOI: 10.1089/ars.2024.0674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Significance: Cholesterol plays a crucial role in the brain, where it is highly concentrated and tightly regulated to support normal brain functions. It serves as a vital component of cell membranes, ensuring their integrity, and acts as a key regulator of various brain processes. Dysregulation of cholesterol metabolism in the brain has been linked to impaired brain function and the onset of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and Huntington's disease. Recent Advances: A significant advancement has been the identification of astrocyte-derived apoliprotein E as a key regulator of de novo cholesterol biosynthesis in neurons, providing insights into how extracellular signals influence neuronal cholesterol levels. In addition, the development of antibody-based therapies, particularly for AD, presents promising opportunities for therapeutic interventions. Critical Issues: Despite significant research, the association between cholesterol and neurodegenerative diseases remains inconclusive. It is crucial to distinguish between plasma cholesterol and brain cholesterol, as these pools are relatively independent. This differentiation should be considered when evaluating statin-based treatment approaches. Furthermore, assessing not only the total cholesterol content in the brain but also its distribution among different types of brain cells is essential. Future Direction: Establishing a causal link between changes in brain/plasma cholesterol levels and the onset of brain dysfunction/neurodegenerative diseases remains a key objective. In addition, conducting cell-specific analyses of cholesterol homeostasis in various types of brain cells under pathological conditions will enhance our understanding of cholesterol metabolism in neurodegenerative diseases. Manipulating cholesterol levels to restore homeostasis may represent a novel approach for alleviating neurological symptoms. Antioxid. Redox Signal. 41, 1051-1072.
Collapse
Affiliation(s)
- Keqiang He
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiang Liu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| |
Collapse
|
3
|
Zhang J, Zeng L, Bu L, Liao H, Wang M, Xiong Y, Cao F. Association between high uric acid and the risk of Parkinson's disease: A meta-analysis. Medicine (Baltimore) 2024; 103:e38947. [PMID: 39058857 PMCID: PMC11272381 DOI: 10.1097/md.0000000000038947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Uric acid, as an important antioxidant substance in human body, has attracted much attention in relation to the risk of Parkinson's disease (PD). However, the causal relationship between them is still controversial. We perform a meta-analysis to summarize the available evidence from cohort studies on the association between high uric acid and the risk of PD. METHODS We searched the Cochrane Library, PubMed, Medline, and Embase to obtain the Odds Ratio (OR) of high uric acid and PD and pooled the data using RevMan software (v5.4; Cochrane library). RESULTS A total of 18 studies involving more than 840,774 participants were included. Overall, we found a significant association (OR = 0.84; 95% CI: 0.77-0.91) between high uric acid and PD. Subgroup analysis was stratified by gender, indicating more statistically significant protective effects of serum urate in men (OR = 0.66; 95% CI: 0.54-0.81) than that of in women (OR = 0.86; 95% CI: 0.76-0.98). People under the age of 60 (OR = 0.53, 95% CI: 0.33-0.86) are more likely to benefit from high uric acid than people over age of 60 (OR = 0.73, 95% CI: 0.63-0.86). The resistance of high uric acid to PD in LRRK2 mutation carriers (OR = 0.22, 95% CI: 0.11-0.45) is stronger than that in non-manifesting LRRK2 mutation carriers (OR = 0.37, 95% CI: 0.16-0.85). In addition, a dose-response trend of serum urate to reduce PD risk was also observed (OR = 0.68; 95% CI: 0.48-0.93). CONCLUSION Our study confirms a significant association between high uric acid and the risk of PD, especially in men under 60 years old, and a dose-response trend of uric acid to reduce PD risk was also observed. Furthermore, LRRK2 mutation carriers are more likely to benefit from high uric acid than non-manifesting LRRK2 mutation carriers.
Collapse
Affiliation(s)
- Jieyu Zhang
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| | - Longhai Zeng
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| | - Lufang Bu
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| | - Hairong Liao
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| | - Meixiang Wang
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| | - Yan Xiong
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| | - Fei Cao
- Fuzhou Medical Department of Nanchang University, Fuzhou, China
| |
Collapse
|
4
|
Cahill CM, Sarang SS, Bakshi R, Xia N, Lahiri DK, Rogers JT. Neuroprotective Strategies and Cell-Based Biomarkers for Manganese-Induced Toxicity in Human Neuroblastoma (SH-SY5Y) Cells. Biomolecules 2024; 14:647. [PMID: 38927051 PMCID: PMC11201412 DOI: 10.3390/biom14060647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/24/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Manganese (Mn) is an essential heavy metal in the human body, while excess Mn leads to neurotoxicity, as observed in this study, where 100 µM of Mn was administered to the human neuroblastoma (SH-SY5Y) cell model of dopaminergic neurons in neurodegenerative diseases. We quantitated pathway and gene changes in homeostatic cell-based adaptations to Mn exposure. Utilizing the Gene Expression Omnibus, we accessed the GSE70845 dataset as a microarray of SH-SY5Y cells published by Gandhi et al. (2018) and applied statistical significance cutoffs at p < 0.05. We report 74 pathway and 10 gene changes with statistical significance. ReactomeGSA analyses demonstrated upregulation of histones (5 out of 10 induced genes) and histone deacetylases as a neuroprotective response to remodel/mitigate Mn-induced DNA/chromatin damage. Neurodegenerative-associated pathway changes occurred. NF-κB signaled protective responses via Sirtuin-1 to reduce neuroinflammation. Critically, Mn activated three pathways implicating deficits in purine metabolism. Therefore, we validated that urate, a purine and antioxidant, mitigated Mn-losses of viability in SH-SY5Y cells. We discuss Mn as a hypoxia mimetic and trans-activator of HIF-1α, the central trans-activator of vascular hypoxic mitochondrial dysfunction. Mn induced a 3-fold increase in mRNA levels for antioxidant metallothionein-III, which was induced 100-fold by hypoxia mimetics deferoxamine and zinc.
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Sanjan S. Sarang
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Rachit Bakshi
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Ning Xia
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| | - Debomoy K. Lahiri
- Department of Psychiatry and Medical & Molecular Genetics, Indiana Alzheimer’s Disease Research Center, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Jack T. Rogers
- Neurochemistry Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; (C.M.C.); (S.S.S.); (R.B.); (N.X.)
| |
Collapse
|
5
|
Ngo HKC, Le H, Ayer SJ, Crotty GF, Schwarzschild MA, Bakshi R. Short-term lipopolysaccharide treatment leads to astrocyte activation in LRRK2 G2019S knock-in mice without loss of dopaminergic neurons. RESEARCH SQUARE 2024:rs.3.rs-4076333. [PMID: 38562908 PMCID: PMC10984011 DOI: 10.21203/rs.3.rs-4076333/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background The G2019S mutation of LRRK2, which enhances kinase activity of the protein, confers a substantial risk of developing Parkinson's disease (PD). However, the mutation demonstrates incomplete penetrance, suggesting the involvement of other genetic or environmental modulating factors. Here, we investigated whether LRRK2 G2019S knock-in (KI) mice treated with the inflammogen lipopolysaccharide (LPS) could model LRRK2 PD. Results We found that short-term (2 weeks) treatment with LPS did not result in the loss of dopaminergic neurons in either LRRK2 G2019S KI or wild-type (WT) mice. Compared with WT mice, LRRK2 G2019S-KI mice showed incomplete recovery from LPS-induced weight loss. In LRRK2 G2019S KI mice, LPS treatment led to upregulated phosphorylation of LRRK2 at the autophosphorylation site Serine 1292, which is known as a direct readout of LRRK2 kinase activity. LPS treatment caused a greater increase in the activated astrocyte marker glial fibrillary acidic protein (GFAP) in the striatum and substantia nigra of LRRK2 G2019S mice than in those of WT mice. The administration of caffeine, which was recently identified as a biomarker of resistance to developing PD in individuals with LRRK2 mutations, attenuated LPS-induced astrocyte activation specifically in LRRK2 G2019S KI mice. Conclusions Our findings suggest that 2 weeks of exposure to LPS is not sufficient to cause dopaminergic neuronal loss in LRRK2 G2019S KI mice but rather results in increased astrocyte activation, which can be ameliorated by caffeine.
Collapse
|
6
|
Korczowska-Łącka I, Słowikowski B, Piekut T, Hurła M, Banaszek N, Szymanowicz O, Jagodziński PP, Kozubski W, Permoda-Pachuta A, Dorszewska J. Disorders of Endogenous and Exogenous Antioxidants in Neurological Diseases. Antioxidants (Basel) 2023; 12:1811. [PMID: 37891890 PMCID: PMC10604347 DOI: 10.3390/antiox12101811] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In diseases of the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), stroke, amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and even epilepsy and migraine, oxidative stress load commonly surpasses endogenous antioxidative capacity. While oxidative processes have been robustly implicated in the pathogenesis of these diseases, the significance of particular antioxidants, both endogenous and especially exogenous, in maintaining redox homeostasis requires further research. Among endogenous antioxidants, enzymes such as catalase, superoxide dismutase, and glutathione peroxidase are central to disabling free radicals, thereby preventing oxidative damage to cellular lipids, proteins, and nucleic acids. Whether supplementation with endogenously occurring antioxidant compounds such as melatonin and glutathione carries any benefit, however, remains equivocal. Similarly, while the health benefits of certain exogenous antioxidants, including ascorbic acid (vitamin C), carotenoids, polyphenols, sulforaphanes, and anthocyanins are commonly touted, their clinical efficacy and effectiveness in particular neurological disease contexts need to be more robustly defined. Here, we review the current literature on the cellular mechanisms mitigating oxidative stress and comment on the possible benefit of the most common exogenous antioxidants in diseases such as AD, PD, ALS, HD, stroke, epilepsy, and migraine. We selected common neurological diseases of a basically neurodegenerative nature.
Collapse
Affiliation(s)
- Izabela Korczowska-Łącka
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Thomas Piekut
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Agnieszka Permoda-Pachuta
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| |
Collapse
|
7
|
Young JE, Wu M, Hunsberger HC. Editorial: Sex and gender differences in neurodegenerative diseases. Front Neurosci 2023; 17:1175674. [PMID: 37008208 PMCID: PMC10061136 DOI: 10.3389/fnins.2023.1175674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Affiliation(s)
- Jessica Elaine Young
- Laboratory Medicine and Pathology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Holly C. Hunsberger
- Department of Neuroscience, Center for Neurodegenerative Diseases and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Department of Foundational Sciences and Humanities, The Chicago Medical School, North Chicago, IL, United States
| |
Collapse
|
8
|
Who is at Risk of Parkinson Disease? Refining the Preclinical Phase of GBA1 and LRRK2 Variant Carriers: a Clinical, Biochemical, and Imaging Approach. Curr Neurol Neurosci Rep 2023; 23:121-130. [PMID: 36881256 PMCID: PMC10119235 DOI: 10.1007/s11910-023-01259-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE OF REVIEW Genetic variants in GBA1 and LRRK2 genes are the commonest genetic risk factor for Parkinson disease (PD); however, the preclinical profile of GBA1 and LRRK2 variant carriers who will develop PD is unclear. This review aims to highlight the more sensitive markers that can stratify PD risk in non-manifesting GBA1 and LRRK2 variant carriers. RECENT FINDINGS Several case-control and a few longitudinal studies evaluated clinical, biochemical, and neuroimaging markers within cohorts of non-manifesting carriers of GBA1 and LRRK2 variants. Despite similar levels of penetrance of PD in GBA1 and LRRK2 variant carriers (10-30%), these individuals have distinct preclinical profiles. GBA1 variant carriers at higher risk of PD can present with prodromal symptoms suggestive of PD (hyposmia), display increased α-synuclein levels in peripheral blood mononuclear cells, and show dopamine transporter abnormalities. LRRK2 variant carriers at higher risk of PD might show subtle motor abnormalities, but no prodromal symptoms, higher exposure to some environmental factors (non-steroid anti-inflammatory drugs), and peripheral inflammatory profile. This information will help clinicians tailor appropriate screening tests and counseling and facilitate researchers in the development of predictive markers, disease-modifying treatments, and selection of healthy individuals who might benefit from preventive interventions.
Collapse
|
9
|
Koros C, Simitsi AM, Papagiannakis N, Bougea A, Prentakis A, Papadimitriou D, Pachi I, Beratis I, Stanitsa E, Angelopoulou E, Antonelou R, Bregianni M, Lourentzos K, Papageorgiou SG, Bonakis A, Trapali XG, Stamelou M, Stefanis L. Serum Uric Acid as a Putative Biomarker in Prodromal Parkinson's Disease: Longitudinal Data from the PPMI Study. JOURNAL OF PARKINSON'S DISEASE 2023; 13:811-818. [PMID: 37424476 PMCID: PMC10473106 DOI: 10.3233/jpd-230007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/01/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND The role of blood uric acid as a biomarker in symptomatic motor PD has been increasingly established in the literature. OBJECTIVE Our present study assessed the role of serum uric acid as a putative biomarker in a prodromal PD cohort [REM Sleep Behavior disorder (RBD) and Hyposmia] followed longitudinally. METHODS Longitudinal 5-year serum uric acid measurement data of 39 RBD patients and 26 Hyposmia patients with an abnormal DATSCAN imaging were downloaded from the Parkinson's Progression Markers Initiative database. These cohorts were compared with 423 de novo PD patients and 196 healthy controls enrolled in the same study. RESULTS After adjusting for age, sex, body mass index, and concomitant disorders (hypertension/gout), baseline and longitudinal serum uric acid levels were higher in the RBD subgroup as compared to the established PD cohort (p = 0.004 and p = 0.001). (Baseline RBD 6.07±1.6 vs. Baseline PD 5.35±1.3 mg/dL and Year-5 RBD 5.7±1.3 vs. Year-5 PD 5.26±1.33). This was also true for longitudinal measurements in the Hyposmic subgroup (p = 0.008) (Baseline Hyposmic 5.7±1.6 vs. PD 5.35±1.3 mg/dL and Year-5 Hyposmic 5.58±1.6 vs. PD 5.26±1.33). CONCLUSION Our results indicate that serum uric acid levels are higher in prodromal PD subjects with ongoing dopaminergic degeneration compared to those with manifest PD. These data indicate that the well-established decrease in the levels of serum uric acid occurs with the transition from prodromal to clinical PD. Whether the higher levels of serum uric acid observed in prodromal PD may provide protection against conversion to full-blown clinical PD will require further study.
Collapse
Affiliation(s)
- Christos Koros
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina-Maria Simitsi
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Papagiannakis
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Bougea
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Prentakis
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Nuclear Medicine Unit, Attikon Hospital, Athens, Greece
| | | | - Ioanna Pachi
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ion Beratis
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Stanitsa
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthalia Angelopoulou
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Roubina Antonelou
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianna Bregianni
- 2 Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Lourentzos
- 2 Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sokratis G. Papageorgiou
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Bonakis
- 2 Department of Neurology, Attikon Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Stamelou
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
- Neurology Clinic, Philipps University, Marburg, Germany
- Parkinsons disease and Movement Disorders Dept., HYGEIA Hospital, Athens, Greece
| | - Leonidas Stefanis
- 1 Department of Neurology, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Mozafar M, Kazemian S, Hoseini E, Mohammadi M, Alimoghadam R, Shafie M, Mayeli M. The glucocerebrosidase mutations and uric acid levels in Parkinson's disease: A 3-years investigation of a potential biomarker". Clin Park Relat Disord 2022; 8:100177. [PMID: 36590455 PMCID: PMC9798165 DOI: 10.1016/j.prdoa.2022.100177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/16/2022] [Accepted: 11/13/2022] [Indexed: 12/23/2022] Open
Abstract
Background Blood uric acid level indicates an emerging biomarker in Parkinson's disease (PD). This study aimed to evaluate longitudinal uric acid levels among different kinds of glucocerebrosidase (GBA) mutations and to compare it among sporadic PD, genetic cohort Parkinson's disease (GENPD), genetic cohort unaffected (GENUN), and healthy control (HC) patients. Methods We conducted a study on 654 individuals from the Parkinson's progression markers initiative (PPMI) database. Baseline characteristics, uric acid levels, movement disorder society unified Parkinson's disease rating scale III (MDS-UPDRS III), Hoehn and Yahr Parkinson stage (H&Y stage), and DaT scan specific binding ratio (SBR) data were obtained. Different GBA mutations were collected and categorized into three groups. Longitudinal measurements of uric acid and MDS-UPDRS III score were evaluated during 3-years of follow-up. Result GENPD cohort exhibited a greater MDS-UPDRS III score, H&Y stage, and lower SBR in the right caudate, left caudate, and right putamen compared to sporadic PD. Baseline uric acid level was similar among all groups and different GBA variants. After adjustment for age, sex, and body mass index, the uric acid level was significantly lower in the GENPD group than in HC during year 2 (P-value: 0.009). No significant longitudinal differences were detected for the MDS-UPDRS III score and three groups of GBA mutations. Conclusion This is the first study to assess uric acid levels and MDS-UPDRS III scores among different GBA mutation variants within 3 years of follow-up. We found similar clinical characteristics among different subtypes of GBA mutations.
Collapse
Affiliation(s)
- Mehrdad Mozafar
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Kazemian
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran,Cardiac Primary Prevention Research Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Hoseini
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran,Medical Imaging Department, AMT School, Isfahan Medical Sciences University, Isfahan, Iran
| | - Mohammad Mohammadi
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Rojina Alimoghadam
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahan Shafie
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mayeli
- NeuroTRACT Association, Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran,Iranian Center of Neurological Research, Imam Khomeini Hospital Complex, Tehran, Iran,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran,Corresponding author.
| | | |
Collapse
|
11
|
Periñán MT, Brolin K, Bandres‐Ciga S, Blauwendraat C, Klein C, Gan‐Or Z, Singleton A, Gomez‐Garre P, Swanberg M, Mir P, Noyce A. Effect Modification between Genes and Environment and Parkinson's Disease Risk. Ann Neurol 2022; 92:715-724. [PMID: 35913124 PMCID: PMC9588606 DOI: 10.1002/ana.26467] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative condition in which genetic and environmental factors interact to contribute to its etiology. Remarkable progress has been made in deciphering disease etiology through genetic approaches, but there is limited data about how environmental and genetic factors interact to modify penetrance, risk, and disease severity. Here, we provide insights into environmental modifiers of PD, discussing precedents from other neurological and non-neurological conditions. Based on these examples, we outline genetic and environmental factors contributing to PD and review potential environmental modifiers of penetrance and clinical variability in monogenic and idiopathic PD. We also highlight the potential challenges and propose how future studies might tackle these important questions. ANN NEUROL 2022;92:715-724.
Collapse
Affiliation(s)
- Maria Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaMadridSpain
| | - Kajsa Brolin
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Department of Experimental Medical ScienceLund UniversityLundSweden
| | - Sara Bandres‐Ciga
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Christine Klein
- Institute of Neurogenetics and Department of NeurologyUniversity of Lübeck and University Hospital Schleswig‐HolsteinLübeckGermany
| | - Ziv Gan‐Or
- The Neuro (Montreal Neurological Institute‐Hospital)McGill UniversityMontrealQuebecCanada,Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada,Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| | - Andrew Singleton
- Laboratory of Neurogenetics, Molecular Genetics Section, National Institute on AgingNational Institutes of HealthBethesdaMarylandUSA
| | - Pilar Gomez‐Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaMadridSpain
| | - Maria Swanberg
- Translational Neurogenetics Unit, Wallenberg Neuroscience Center, Department of Experimental Medical ScienceLund UniversityLundSweden
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de SevillaHospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaMadridSpain
| | - Alastair Noyce
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK,Preventive Neurology Unit, Centre for Prevention, Detection and Diagnosis, Wolfson Institute of Population HealthQueen Mary University of LondonLondonUK
| |
Collapse
|
12
|
Fazlollahi A, Zahmatyar M, Alizadeh H, Noori M, Jafari N, Nejadghaderi SA, Sullman MJM, Gharagozli K, Kolahi AA, Safiri S. Association between gout and the development of Parkinson's disease: a systematic review and meta-analysis. BMC Neurol 2022; 22:383. [PMID: 36221048 PMCID: PMC9552480 DOI: 10.1186/s12883-022-02874-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND As a natural antioxidant, uric acid plays a protective role against neurodegenerative disorders, including Parkinson's disease (PD). Therefore, the risk of PD has been found to be lower in people with hyperuricemia. In this article, we conducted a systematic review and meta-analysis to investigate whether gout affects the future risk of developing PD. METHODS We searched PubMed, Scopus, the Web of Science, and Google Scholar to find relevant studies, up to March 16, 2022. Studies investigating the risk of PD, following a gout diagnosis, were included if they were cross-sectional, case-control or cohort studies. The Newcastle Ottawa Scale (NOS) checklist was used to assess the quality of all included studies. The meta-analysis was performed using STATA 17.0. RESULTS Ten studies were included, which were comprised of three case-controls, six cohort studies and one nested case-control study. We found no significant association between gout and the risk of PD among both sexes (RR = 0.94, 95% CI: 0.86-1.04), although the association was significant for females (RR = 1.09; 95% CI: 1.02-1.17). Subgroup analysis also showed no significant findings by age group, whether they were receiving treatment for gout, study design, quality assessment score, and method of gout ascertainment. In contrast, the studies that defined PD according to the use of drugs showed significant results (RR = 0.82; 95% CI: 0.76-0.89). There was a significant publication bias on the association between gout and PD. CONCLUSIONS The presence of gout had no significant effect on the risk of subsequently developing PD. Further analyses are recommended to investigate the effects of demographic and behavioral risk factors.
Collapse
Affiliation(s)
- Asra Fazlollahi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Zahmatyar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Alizadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Noori
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Jafari
- Department of Epidemiology and Biostatistics, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Aria Nejadghaderi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Koroush Gharagozli
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Niotis K, West AB, Saunders-Pullman R. Who to Enroll in Parkinson Disease Prevention Trials? The Case for Genetically At-Risk Cohorts. Neurology 2022; 99:10-18. [PMID: 35970585 DOI: 10.1212/wnl.0000000000200812] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/15/2022] [Indexed: 12/15/2022] Open
Abstract
Therapies that prevent the occurrence of Parkinson disease (PD) (primary prevention) or mitigate the progression of symptoms in those with early disease (secondary prevention) are a critical unmet need in disease management. Despite great promise, PD prevention trials have not yet demonstrated success. Initiation of treatment too late in the disease course and the heterogeneity of disease are obstacles that may have contributed to the failure. Genetically stratified groups offer many advantages to primary and secondary prevention trials. In addition to their ease of identification, they decrease disease heterogeneity on several levels. Particularly, they comprise a phenotypically and pathologically enriched group with defined clinical features, pathogenic mechanisms and associated proteins that may serve as specific trial endpoints, therapeutic targets and biomarkers for disease state, and pharmacodynamic and pharmacokinetic status. However, challenges arise from genetic variant heterogeneity, from reduced penetrance whereby many carriers will not develop PD, and in recruiting a population that will meet the desired outcome in the proposed study duration. In this review, we discussed the opportunities afforded by the enrollment of genetically stratified cohorts (i.e., leucine-rich repeat kinase 2 and glucocerebrosidase 1) into prevention trials with a primary focus on primary prevention trials. We also outlined challenges surrounding the enrollment of these cohorts and offered suggestions to leverage their many advantages.
Collapse
Affiliation(s)
- Kellyann Niotis
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC
| | - Andrew B West
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC
| | - Rachel Saunders-Pullman
- From the Department of Neurology (K.N., R.S.-P.), Mount Sinai Beth Israel Medical Center; Department of Neurology (K.N., R.S.-P.), Icahn School of Medicine at Mount Sinai, New York; and Duke Center for Neurodegeneration Research (A.B.W.), Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC.
| |
Collapse
|
14
|
Trinh J, Schymanski EL, Smajic S, Kasten M, Sammler E, Grünewald A. Molecular mechanisms defining penetrance of LRRK2-associated Parkinson's disease. MED GENET-BERLIN 2022; 34:103-116. [PMID: 38835904 PMCID: PMC11006382 DOI: 10.1515/medgen-2022-2127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Mutations in Leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of dominantly inherited Parkinson's disease (PD). LRRK2 mutations, among which p.G2019S is the most frequent, are inherited with reduced penetrance. Interestingly, the disease risk associated with LRRK2 G2019S can vary dramatically depending on the ethnic background of the carrier. While this would suggest a genetic component in the definition of LRRK2-PD penetrance, only few variants have been shown to modify the age at onset of patients harbouring LRRK2 mutations, and the exact cellular pathways controlling the transition from a healthy to a diseased state currently remain elusive. In light of this knowledge gap, recent studies also explored environmental and lifestyle factors as potential modifiers of LRRK2-PD. In this article, we (i) describe the clinical characteristics of LRRK2 mutation carriers, (ii) review known genes linked to LRRK2-PD onset and (iii) summarize the cellular functions of LRRK2 with particular emphasis on potential penetrance-related molecular mechanisms. This section covers LRRK2's involvement in Rab GTPase and immune signalling as well as in the regulation of mitochondrial homeostasis and dynamics. Additionally, we explored the literature with regard to (iv) lifestyle and (v) environmental factors that may influence the penetrance of LRRK2 mutations, with a view towards further exposomics studies. Finally, based on this comprehensive overview, we propose potential future in vivo, in vitro and in silico studies that could provide a better understanding of the processes triggering PD in individuals with LRRK2 mutations.
Collapse
Affiliation(s)
- Joanne Trinh
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Emma L. Schymanski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Semra Smajic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Meike Kasten
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Esther Sammler
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Department of Neurology, School of Medicine, Dundee, Ninewells Hospital, Dundee, UK
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
15
|
Seibler P, Rakovic A. Patient-derived cells - an irreplaceable tool for research of reduced penetrance in movement disorders. MED GENET-BERLIN 2022; 34:125-130. [PMID: 38835901 PMCID: PMC11006347 DOI: 10.1515/medgen-2022-2133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Movement disorders comprise a clinically, pathologically, and genetically heterogeneous group of diseases associated with the phenomenon of reduced penetrance. Penetrance refers to the likelihood that a clinical condition will occur when a particular genotype is present. Elucidating the cause of reduced penetrance may contribute to more personalized medicine by identifying genetic factors that may prevent individuals from developing disease. Therefore, patient material becomes an irreplaceable resource in this approach. It is needed to identify genetic modifiers of the disease in the first place and to subsequently elucidate underlying mechanisms in endogenous human cell models that provide the entire genetic background.
Collapse
Affiliation(s)
- Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | |
Collapse
|
16
|
Wang P, Cui P, Luo Q, Chen J, Tang H, Zhang L, Chen S, Ma J. Penetrance of Parkinson disease LRRK2 G2385R-associated variant in the Chinese population. Eur J Neurol 2022; 29:2639-2644. [PMID: 35608967 DOI: 10.1111/ene.15417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE Penetrance estimates of the leucine-rich repeat kinase 2 (LRRK2) variants for Parkinson disease (PD) vary widely. G2385R is one of the most common LRRK2 variants in Asian populations, and its penetrance is currently unknown. We aimed to estimate the penetrance of G2385R in the Chinese population. METHODS The G2385R variant was tested by Sanger sequencing in 6386 participants older than 50 years, all from the community cohort established by Shanghai Ruijin Hospital in 2009-2011. G2385R carriers and matched noncarriers underwent a brief questionnaire survey (including sex, current age, PD diagnosis, and age at onset) and face-to-face PD assessment during 2020-2021. The penetrance of PD was estimated by the Kaplan-Meier method. RESULTS A total of 396 G2385R carriers and 415 noncarriers were included, after excluding those with a baseline diagnosis of PD or unwilling to participate. In G2385R carriers, the penetrance of PD was 1.64% at 70 years, 10.26% at 80 years, and 18.49% at 90 years, and reached 25.90% at 95 years. The penetrance of PD in G2385R carriers was higher than in noncarriers (p = 0.0071). In noncarriers, only 0%, 3.72%, and 9.66% developed parkinsonism by 70, 80, and 90 years of age. Among carriers and noncarriers, there were no statistically significant differences in penetrance comparisons between males and females, or between urban and rural. CONCLUSIONS The lifetime penetrance of LRRK2 G2385R in the Chinese population was 25.9%. The penetrance modifier of G2385R in our study was age-related. Further investigation of genetic and environmental modifiers affecting G2385R penetrance is warranted.
Collapse
Affiliation(s)
- Pei Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijing Cui
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Luo
- Department of Pediatric Hematology-Oncology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Chen
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huidong Tang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lina Zhang
- Department of Biostatistics, Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfang Ma
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Manini A, Abati E, Comi GP, Corti S, Ronchi D. Mitochondrial DNA homeostasis impairment and dopaminergic dysfunction: A trembling balance. Ageing Res Rev 2022; 76:101578. [PMID: 35114397 DOI: 10.1016/j.arr.2022.101578] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/26/2021] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Maintenance of mitochondrial DNA (mtDNA) homeostasis includes a variety of processes, such as mtDNA replication, repair, and nucleotides synthesis, aimed at preserving the structural and functional integrity of mtDNA molecules. Mutations in several nuclear genes (i.e., POLG, POLG2, TWNK, OPA1, DGUOK, MPV17, TYMP) impair mtDNA maintenance, leading to clinical syndromes characterized by mtDNA depletion and/or deletions in affected tissues. In the past decades, studies have demonstrated a progressive accumulation of multiple mtDNA deletions in dopaminergic neurons of the substantia nigra in elderly population and, to a greater extent, in Parkinson's disease patients. Moreover, parkinsonism has been frequently described as a prominent clinical feature in mtDNA instability syndromes. Among Parkinson's disease-related genes with a significant role in mitochondrial biology, PARK2 and LRRK2 specifically take part in mtDNA maintenance. Moreover, a variety of murine models (i.e., "Mutator", "MitoPark", "PD-mitoPstI", "Deletor", "Twinkle-dup" and "TwinkPark") provided in vivo evidence that mtDNA stability is required to preserve nigrostriatal integrity. Here, we review and discuss the clinical, genetic, and pathological background underlining the link between impaired mtDNA homeostasis and dopaminergic degeneration.
Collapse
|
18
|
Serum Uric Acid Levels in Parkinson’s Disease: A Cross-Sectional Electronic Medical Record Database Study from a Tertiary Referral Centre in Romania. Medicina (B Aires) 2022; 58:medicina58020245. [PMID: 35208569 PMCID: PMC8877142 DOI: 10.3390/medicina58020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background and Objectives: Parkinson’s disease (PD) is a prevalent neurodegenerative condition responsible for progressive motor and non-motor symptoms. Currently, no prophylactic or disease-modifying interventions are available. Uric acid (UA) is a potent endogenous antioxidant, resulting from purine metabolism. It is responsible for about half of the antioxidant capacity of the plasma. Increasing evidence suggests that lower serum UA levels are associated with an increased risk of developing PD and with faster disease progression. Materials and Methods: We conducted an electronic medical record database study to investigate the associations between UA levels and different characteristics of PD. Results: Out of 274 datasets from distinct patients with PD, 49 complied with the predefined inclusion and exclusion criteria. Lower UA levels were significantly associated with the severity of parkinsonism according to the Hoehn and Yahr stage (rs = 0.488, p = 0.002), with the motor complications of long-term dopaminergic treatment (r = 0.333, p = 0.027), and with the presence of neurocognitive impairment (r = 0.346, p = 0.021). Conclusions: Oxidative stress is considered a key player in the etiopathogenesis of PD, therefore the involvement of lower UA levels in the development and progression of PD is plausible. Data on the potential therapeutic roles of elevating serum UA (e.g., by precursor administration or diet manipulation) are scarce, but considering the accumulating epidemiological evidence, the topic warrants further research.
Collapse
|
19
|
Plasma Metabolite Signature Classifies Male LRRK2 Parkinson’s Disease Patients. Metabolites 2022; 12:metabo12020149. [PMID: 35208223 PMCID: PMC8876175 DOI: 10.3390/metabo12020149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease, causing loss of motor and nonmotor function. Diagnosis is based on clinical symptoms that do not develop until late in the disease progression, at which point the majority of the patients’ dopaminergic neurons are already destroyed. While many PD cases are idiopathic, hereditable genetic risks have been identified, including mutations in the gene for LRRK2, a multidomain kinase with roles in autophagy, mitochondrial function, transcription, molecular structural integrity, the endo-lysosomal system, and the immune response. A definitive PD diagnosis can only be made post-mortem, and no noninvasive or blood-based disease biomarkers are currently available. Alterations in metabolites have been identified in PD patients, suggesting that metabolomics may hold promise for PD diagnostic tools. In this study, we sought to identify metabolic markers of PD in plasma. Using a 1H-13C heteronuclear single quantum coherence spectroscopy (HSQC) NMR spectroscopy metabolomics platform coupled with machine learning (ML), we measured plasma metabolites from approximately age/sex-matched PD patients with G2019S LRRK2 mutations and non-PD controls. Based on the differential level of known and unknown metabolites, we were able to build a ML model and develop a Biomarker of Response (BoR) score, which classified male LRRK2 PD patients with 79.7% accuracy, 81.3% sensitivity, and 78.6% specificity. The high accuracy of the BoR score suggests that the metabolomics/ML workflow described here could be further utilized in the development of a confirmatory diagnostic for PD in larger patient cohorts. A diagnostic assay for PD will aid clinicians and their patients to quickly move toward a definitive diagnosis, and ultimately empower future clinical trials and treatment options.
Collapse
|
20
|
Boas SM, Joyce KL, Cowell RM. The NRF2-Dependent Transcriptional Regulation of Antioxidant Defense Pathways: Relevance for Cell Type-Specific Vulnerability to Neurodegeneration and Therapeutic Intervention. Antioxidants (Basel) 2021; 11:antiox11010008. [PMID: 35052512 PMCID: PMC8772787 DOI: 10.3390/antiox11010008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress has been implicated in the etiology and pathobiology of various neurodegenerative diseases. At baseline, the cells of the nervous system have the capability to regulate the genes for antioxidant defenses by engaging nuclear factor erythroid 2 (NFE2/NRF)-dependent transcriptional mechanisms, and a number of strategies have been proposed to activate these pathways to promote neuroprotection. Here, we briefly review the biology of the transcription factors of the NFE2/NRF family in the brain and provide evidence for the differential cellular localization of NFE2/NRF family members in the cells of the nervous system. We then discuss these findings in the context of the oxidative stress observed in two neurodegenerative diseases, Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), and present current strategies for activating NFE2/NRF-dependent transcription. Based on the expression of the NFE2/NRF family members in restricted populations of neurons and glia, we propose that, when designing strategies to engage these pathways for neuroprotection, the relative contributions of neuronal and non-neuronal cell types to the overall oxidative state of tissue should be considered, as well as the cell types which have the greatest intrinsic capacity for producing antioxidant enzymes.
Collapse
Affiliation(s)
- Stephanie M. Boas
- Department of Neuroscience, Southern Research, 2000 9th Avenue South, Birmingham, AL 35205, USA; (S.M.B.); (K.L.J.)
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294, USA
| | - Kathlene L. Joyce
- Department of Neuroscience, Southern Research, 2000 9th Avenue South, Birmingham, AL 35205, USA; (S.M.B.); (K.L.J.)
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294, USA
| | - Rita M. Cowell
- Department of Neuroscience, Southern Research, 2000 9th Avenue South, Birmingham, AL 35205, USA; (S.M.B.); (K.L.J.)
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294, USA
- Correspondence:
| |
Collapse
|
21
|
Parga JA, Rodriguez-Perez AI, Garcia-Garrote M, Rodriguez-Pallares J, Labandeira-Garcia JL. NRF2 Activation and Downstream Effects: Focus on Parkinson's Disease and Brain Angiotensin. Antioxidants (Basel) 2021; 10:antiox10111649. [PMID: 34829520 PMCID: PMC8614768 DOI: 10.3390/antiox10111649] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are signalling molecules used to regulate cellular metabolism and homeostasis. However, excessive ROS production causes oxidative stress, one of the main mechanisms associated with the origin and progression of neurodegenerative disorders such as Parkinson's disease. NRF2 (Nuclear Factor-Erythroid 2 Like 2) is a transcription factor that orchestrates the cellular response to oxidative stress. The regulation of NRF2 signalling has been shown to be a promising strategy to modulate the progression of the neurodegeneration associated to Parkinson's disease. The NRF2 pathway has been shown to be affected in patients with this disease, and activation of NRF2 has neuroprotective effects in preclinical models, demonstrating the therapeutic potential of this pathway. In this review, we highlight recent advances regarding the regulation of NRF2, including the effect of Angiotensin II as an endogenous signalling molecule able to regulate ROS production and oxidative stress in dopaminergic neurons. The genes regulated and the downstream effects of activation, with special focus on Kruppel Like Factor 9 (KLF9) transcription factor, provide clues about the mechanisms involved in the neurodegenerative process as well as future therapeutic approaches.
Collapse
Affiliation(s)
- Juan A. Parga
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
- Correspondence: (J.A.P.); (J.L.L.-G.)
| | - Ana I. Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Maria Garcia-Garrote
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Jannette Rodriguez-Pallares
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Jose L. Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
- Correspondence: (J.A.P.); (J.L.L.-G.)
| |
Collapse
|
22
|
Olubodun-Obadun TG, Ishola IO, Adeyemi OO. Potentials of autophagy enhancing natural products in the treatment of Parkinson disease. Drug Metab Pers Ther 2021; 0:dmdi-2021-0128. [PMID: 34391219 DOI: 10.1515/dmdi-2021-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/11/2021] [Indexed: 11/15/2022]
Abstract
Parkinson disease (PD) is a progressive neurodegenerative movement disorder characterized by motor and non-motor symptoms due to loss of striatal dopaminergic neurons and disruption of degradation signaling leading to the formation of Lewy bodies (aggregation of α-synuclein). Presently, there are no disease modifying therapy for PD despite improvement in the understanding of the disease pathogenesis. However, the drugs currently used in PD management provide symptomatic relieve for motor symptoms without significant improvement in non-motor complications, thus, a public health burden on caregivers and healthcare systems. There is therefore the need to discover disease modifying therapy with strong potential to halt the disease progression. Recent trend has shown that the dysfunction of lysosomal-autophagy pathway is highly implicated in PD pathology, hence, making autophagy a key player owing to its involvement in degradation and clearance of misfolded α-synuclein (a major hallmark in PD pathology). In this review, we described the current drugs/strategy in the management of PD including targeting the autophagy pathway as a novel approach that could serve as potential intervention for PD management. The discovery of small molecules or natural products capable of enhancing autophagy mechanism could be a promising strategy for PD treatment.
Collapse
Affiliation(s)
- Taiwo G Olubodun-Obadun
- Department of Pharmacology, Therapeutics and Toxicology, College of Medicine, University of Lagos, Lagos, Lagos State, Nigeria
| | - Ismail O Ishola
- Department of Pharmacology, Therapeutics and Toxicology, College of Medicine, University of Lagos, Lagos, Lagos State, Nigeria
| | - Olufunmilayo O Adeyemi
- Department of Pharmacology, Therapeutics and Toxicology, College of Medicine, University of Lagos, Lagos, Lagos State, Nigeria
| |
Collapse
|
23
|
Olubodun-Obadun TG, Ishola IO, Adeyemi OO. Potentials of autophagy enhancing natural products in the treatment of Parkinson disease. Drug Metab Pers Ther 2021; 37:99-110. [PMID: 35737301 DOI: 10.1515/dmpt-2021-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/11/2021] [Indexed: 06/15/2023]
Abstract
Parkinson disease (PD) is a progressive neurodegenerative movement disorder characterized by motor and non-motor symptoms due to loss of striatal dopaminergic neurons and disruption of degradation signaling leading to the formation of Lewy bodies (aggregation of α-synuclein). Presently, there are no disease modifying therapy for PD despite improvement in the understanding of the disease pathogenesis. However, the drugs currently used in PD management provide symptomatic relieve for motor symptoms without significant improvement in non-motor complications, thus, a public health burden on caregivers and healthcare systems. There is therefore the need to discover disease modifying therapy with strong potential to halt the disease progression. Recent trend has shown that the dysfunction of lysosomal-autophagy pathway is highly implicated in PD pathology, hence, making autophagy a key player owing to its involvement in degradation and clearance of misfolded α-synuclein (a major hallmark in PD pathology). In this review, we described the current drugs/strategy in the management of PD including targeting the autophagy pathway as a novel approach that could serve as potential intervention for PD management. The discovery of small molecules or natural products capable of enhancing autophagy mechanism could be a promising strategy for PD treatment.
Collapse
Affiliation(s)
- Taiwo G Olubodun-Obadun
- Department of Pharmacology, Therapeutics and Toxicology, College of Medicine, University of Lagos, Lagos, Lagos State, Nigeria
| | - Ismail O Ishola
- Department of Pharmacology, Therapeutics and Toxicology, College of Medicine, University of Lagos, Lagos, Lagos State, Nigeria
| | - Olufunmilayo O Adeyemi
- Department of Pharmacology, Therapeutics and Toxicology, College of Medicine, University of Lagos, Lagos, Lagos State, Nigeria
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Purines have several important physiological functions as part of nucleic acids and as intracellular and extracellular signaling molecules. Purine metabolites, particularly uric acid, have been implicated in congenital and complex diseases. However, their role in complex diseases is not clear and they have both beneficial and detrimental effects on disease pathogenesis. In addition, the relationship between purines and complex diseases is affected by genetic and nutritional factors. This review presents latest findings about the relationship between purines and complex diseases and the effect of genes and nutrients on this relationship. RECENT FINDINGS Evidence from recent studies show strong role of purines in complex diseases. Although they are causal in only few diseases, our knowledge about their role in other diseases is still evolving. Of all the purines, uric acid is the most studied. Uric acid acts as an antioxidant as well as a prooxidant under different conditions, thus, its role in disease also varies. Other purines, adenosine and inosine have been less studied, but they have neuroprotective properties which are valuable in neurodegenerative diseases. SUMMARY Purines are molecules with great potential in disease pathogenesis as either metabolic markers or therapeutic targets. More studies need to be conducted to understand their relevance for complex diseases.
Collapse
Affiliation(s)
- Kendra L Nelson
- Department of Nutrition, Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA
| | | |
Collapse
|
25
|
Bougea A, Koros C, Papagiannakis N, Simitsi AM, Prentakis A, Papadimitriou D, Pachi I, Antonelou R, Angelopoulou E, Beratis I, Bozi M, Papageorgiou SG, Trapali XG, Stamelou M, Stefanis L. Serum Uric Acid in LRRK2 Related Parkinson's Disease: Longitudinal Data from the PPMI Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:633-640. [PMID: 33682725 DOI: 10.3233/jpd-202337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Previous studies have highlighted serum uric acid as a putative idiopathic Parkinson's disease (iPD) biomarker. Only one study, so far, showed higher levels of serum uric acid in leucine-rich repeat kinase 2 (LRRK + 2) carriers compared to those who developed PD, however a longitudinal comparison between LRRK2 + PD and healthy controls (HC) has not been performed. OBJECTIVE The aim of this study was to determine whether there are longitudinal differences in serum uric acid between iPD, LRRK2 + PD and HC and their association with motor and non-motor features. METHODS Longitudinal data of uric acid of 282 de novo iPD, 144 LRRK2 + PD patients, and 195 age-matched HC were obtained from the Parkinson's Progression Markers Initiative (PPMI) database. We also used longitudinal Montreal Cognitive Assessment (MoCA), Movement Disorder Society-Unified Parkinson's Disease Rating Scale part III (MDS-UPDRS-III), Geriatric Depression Scale (GDS) scores, and DaTSCAN striatal binding ratios (SBRs). RESULTS Longitudinal uric acid measurements were significantly lower in LRRK2 + PD patients compared to HC up to 5 years follow-up. There was no significant impact or correlation of adjusted or unadjusted uric acid levels with MoCA, MDS-UPDRS III, or GDS scores, the presence of RBD or DAT-SCAN SBRs. CONCLUSION LRRK2 + PD group had significantly lower uric acid concentrations compared to HC after adjusting for age, sex and baseline BMI up to 5 years follow-up. There were no significant associations between uric acid levels and indices of disease severity. These findings identify serum uric acid as a marker linked to LRRK2 + PD.
Collapse
Affiliation(s)
- Anastasia Bougea
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Koros
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Papagiannakis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina-Maria Simitsi
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Prentakis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Nuclear Medicine Unit, Attikon Hospital, Athens, Greece
| | | | - Ioanna Pachi
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Roubina Antonelou
- 2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthalia Angelopoulou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ion Beratis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Bozi
- 2nd Department of Neurology, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sokratis G Papageorgiou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Stamelou
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Neurology Clinic, Philipps University, Marburg, Germany.,Parkinson's disease and Movement Disorders Department, HYGEIA Hospital, Athens, Greece
| | - Leonidas Stefanis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
26
|
Serum uric acid level as a putative biomarker in Parkinson's disease patients carrying GBA1 mutations: 2-Year data from the PPMI study. Parkinsonism Relat Disord 2021; 84:1-4. [PMID: 33508700 DOI: 10.1016/j.parkreldis.2020.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Blood uric acid represents an important biomarker in sporadic Parkinson's disease (PD). Whether uric acid levels change in genetic forms of PD is beginning to be assessed. The aim of the present study was to evaluate differences in serum uric acid level among PD patients harboring mutations in the glucocerebrosidase (GBA1) gene, sporadic PD, and healthy controls followed longitudinally. METHODS Longitudinal 2-year serum uric acid measurement data of 120 GBA-PD patients have been downloaded from the Parkinson's Progression Markers Initiative (PPMI) database. This cohort was compared with 369 de novo sporadic PD patients and 195 healthy controls enrolled in the same study. RESULTS Following adjustment for age, sex and BMI the GBA-PD cohort exhibited lower 2-year longitudinal uric acid level as compared to the controls (p = 0.016). Baseline uric acid measurements showed only a marginal difference (p = 0.119), but year 2 uric acid levels were lower in the GBA-PD cohort (p < 0.001). There was no difference in baseline, year 2 and 2-year longitudinal serum uric acid in the GBA-PD cohort as compared to sporadic PD (p = 0.664, p = 0.117 and p = 0.315). CONCLUSIONS This is the first study to assess serum uric acid in a GBA-PD cohort. Our findings suggest that low serum uric acid might be a progression biomarker in GBA-PD. However, more studies (ideally longitudinal) on the association between low serum uric acid and clinical data in GBA-PD are needed. These results are consistent with data from previous reports assessing uric acid as a biomarker in other genetic forms of PD.
Collapse
|
27
|
Thaler A, Omer N, Giladi N, Gurevich T, Bar-Shira A, Gana-Weisz M, Goldstein O, Kestenbaum M, Cedarbaum JM, Orr-Urtreger A, Shenhar-Tsarfaty S, Mirelman A. Biochemical markers for severity and risk in GBA and LRRK2 Parkinson's disease. J Neurol 2021; 268:1517-1525. [PMID: 33388928 DOI: 10.1007/s00415-020-10325-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND The phenotype of Parkinson's disease (PD) is variable with mutations in genes such as LRRK2 and GBA explaining part of this heterogeneity. Additional genetic and environmental factors contribute to disease variability. OBJECTIVE To assess the association between biochemical markers, PD severity and probability score for prodromal PD, among GBA and LRRK2 mutation carriers. METHODS Levels of uric acid, vitamin D, C-reactive protein, microalbumin/creatinine ratio (ACR), white blood count (WBC), hemoglobin, platelets, neutrophil/lymphocyte ratio and estimated glomerular filtration rate (eGFR) were assessed from patients with PD and non-manifesting carriers (NMC) of mutations in GBA and LRRK2, together with disease related questionnaires enabling the construction of the MDS prodromal probability score. RESULT A total of 241 patients with PD: 105 idiopathic PD (iPD), 49 LRRK2-PD and 87 GBA-PD and 412 non-manifesting subjects; 74 LRRK2-NMC, 118 GBA-NMC and 220 non-manifesting non-carriers (NMNC), participated in this study. No significant differences in biochemical measures were detected among patients with PD or non-manifesting carriers. Among GBA-PD patients, worse motor performance was associated with ACR (B = 4.68, 95% CI (1.779-7.559); p = 0.002). The probability score for prodromal PD among all non-manifesting participants was associated with eGFR; NMNC (B = - 0.531 95% CI (- 0.879 to - 0.182); p < 0.001, LRRK2-NMC (B = - 1.014 95% CI (- 1.663 to - 0.366); p < 0.001) and GBA-NMC (B = - 0.686 95% CI (1.300 to - 0.071); p = 0.029). CONCLUSION Sub-clinical renal impairment is associated with increased likelihood for prodromal PD regardless of genetic status. While the mechanism behind this finding needs further elucidation, it suggests that kidney function might play a role in PD pathogenesis.
Collapse
Affiliation(s)
- Avner Thaler
- Movement Disorder Unit, Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel-Aviv Medical Center, 6 Weizmann Street, 64239, Tel-Aviv, Israel.
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel.
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel-Aviv Medical Center, Tel Aviv, Israel.
| | - Nurit Omer
- Movement Disorder Unit, Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel-Aviv Medical Center, 6 Weizmann Street, 64239, Tel-Aviv, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Nir Giladi
- Movement Disorder Unit, Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel-Aviv Medical Center, 6 Weizmann Street, 64239, Tel-Aviv, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| | - Tanya Gurevich
- Movement Disorder Unit, Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel-Aviv Medical Center, 6 Weizmann Street, 64239, Tel-Aviv, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| | - Anat Bar-Shira
- Genetic Institute, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - Mali Gana-Weisz
- Genomic Research Laboratory for Neurodegeneration, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - Orly Goldstein
- Genomic Research Laboratory for Neurodegeneration, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - Meir Kestenbaum
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Neurology Department, Meir Medical Center, Kfar Saba, Israel
| | - Jesse M Cedarbaum
- Biogen Inc., Cambridge, MA, USA
- Coeruleus Clinical Sciences LLC, Woodbridge, CT, USA
| | - Avi Orr-Urtreger
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Genomic Research Laboratory for Neurodegeneration, Tel-Aviv Medical Center, Tel Aviv, Israel
| | - Shani Shenhar-Tsarfaty
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Department of Internal Medicine "C", "D", and "E", Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Anat Mirelman
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
- Laboratory of Early Markers of Neurodegeneration, Neurological Institute, Tel-Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
28
|
Neilson LE, Quinn JF, Gray NE. Peripheral Blood NRF2 Expression as a Biomarker in Human Health and Disease. Antioxidants (Basel) 2020; 10:antiox10010028. [PMID: 33396641 PMCID: PMC7824022 DOI: 10.3390/antiox10010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor which plays a critical role in maintenance of cellular redox, has been identified as a therapeutic target in a number of human diseases. Several reports have demonstrated beneficial effects of NRF2 manipulation in animal models of disease, and one NRF2-activating drug, dimethyl fumarate, is already approved for the treatment of multiple sclerosis. However, drug discovery is slowed due to a dearth of biomarkers which can inform target engagement and magnitude and duration of action. Peripheral blood mononuclear cells (PBMCs) are an accessible, minimally-invasive source of biomarkers which can be readily assayed and objectively monitored as a surrogate endpoint of NRF2 activation in clinical trials. We undertook a review of the literature on PBMC NRF2 measurements in human studies to explore its role as a suitable biomarker in various contexts of health and disease. It is clear that NRF2 and its target genes can be readily assayed from PBMCs in multiple disease contexts and may track with disease progression. Further work needs to be undertaken to evaluate its stability but should be considered as an exploratory marker in clinical trials targeting NRF2 activation.
Collapse
Affiliation(s)
- Lee E. Neilson
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (J.F.Q.); (N.E.G.)
- Department of Neurology, Veterans Affairs Medical Center, Portland, OR 97239, USA
- Correspondence: ; Tel.: +1-503-494-7231
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (J.F.Q.); (N.E.G.)
- Department of Neurology, Veterans Affairs Medical Center, Portland, OR 97239, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (J.F.Q.); (N.E.G.)
| |
Collapse
|
29
|
Wen S, Aki T, Unuma K, Uemura K. Chemically Induced Models of Parkinson's Disease: History and Perspectives for the Involvement of Ferroptosis. Front Cell Neurosci 2020; 14:581191. [PMID: 33424553 PMCID: PMC7786020 DOI: 10.3389/fncel.2020.581191] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Ferroptosis is a newly discovered form of necrotic cell death characterized by its dependency on iron and lipid peroxidation. Ferroptosis has attracted much attention recently in the area of neurodegeneration since the involvement of ferroptosis in Parkinson’s disease (PD), a major neurodegenerative disease, has been indicated using animal models. Although PD is associated with both genetic and environmental factors, sporadic forms of PD account for more than 90% of total PD. Following the importance of environmental factors, various neurotoxins are used as chemical inducers of PD both in vivo and in vitro. In contrast to other neurodegenerative diseases such as Alzheimer’s and Huntington’s diseases (AD and HD), many of the characteristics of PD can be reproduced in vivo by the use of specific neurotoxins. Given the indication of ferroptosis in PD pathology, several studies have been conducted to examine whether ferroptosis plays role in the loss of dopaminergic neurons in PD. However, there are still few reports showing an authentic form of ferroptosis in neuronal cells during exposure to the neurotoxins used as PD inducers. In this review article, we summarize the history of the uses of chemicals to create PD models in vivo and in vitro. Besides, we also survey recent reports examining the possible involvement of ferroptosis in chemical models of PD.
Collapse
Affiliation(s)
- Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
30
|
Crotty GF, Lo RY, Schwarzschild MA. If LRRK2 Set the Fire, Can Nonsteroidal Anti-inflammatory Drugs Wet the Flames? Mov Disord 2020; 35:1727-1730. [PMID: 33068466 DOI: 10.1002/mds.28240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Grace F Crotty
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Raymond Y Lo
- Department of Neurology, Hualien Tzu Chi Hospital and Tzu Chi University, Hualien, Taiwan
| | - Michael A Schwarzschild
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Hasimoglu YG, Chen X, Bakshi R, Schwarzschild MA, Macklin EA. Does Serum Urate Change as Parkinson's Disease Progresses? JOURNAL OF PARKINSONS DISEASE 2020; 10:1571-1576. [PMID: 32773396 PMCID: PMC7683051 DOI: 10.3233/jpd-202064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Higher serum urate concentration is associated with decreased risk of Parkinson’s disease (PD) as well as slower disease progression, but its relationship with severity of PD remains unclear. This study investigated whether changes in serum urate concentration over 5 years were associated with disease progression assessed by MDS-UPDRS Part III score, Hoehn and Yahr stage, or DaTscan imaging. Average serum urate concentration was stable over time and change in serum urate concentration did not correlate with worsening of measures of PD progression. These results suggest that serum urate concentration is not a monitoring biomarker of PD progression in early stages.
Collapse
Affiliation(s)
- Yasemin G Hasimoglu
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Xiqun Chen
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Rachit Bakshi
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Michael A Schwarzschild
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, USA
| | - Eric A Macklin
- Department of Medicine, Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
32
|
von Linstow CU, Gan-Or Z, Brundin P. Precision medicine in Parkinson's disease patients with LRRK2 and GBA risk variants - Let's get even more personal. Transl Neurodegener 2020; 9:39. [PMID: 33066808 PMCID: PMC7565766 DOI: 10.1186/s40035-020-00218-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is characterized by motor deficits and a wide variety of non-motor symptoms. The age of onset, rate of disease progression and the precise profile of motor and non-motor symptoms display considerable individual variation. Neuropathologically, the loss of substantia nigra dopaminergic neurons is a key feature of PD. The vast majority of PD patients exhibit alpha-synuclein aggregates in several brain regions, but there is also great variability in the neuropathology between individuals. While the dopamine replacement therapies can reduce motor symptoms, current therapies do not modify the disease progression. Numerous clinical trials using a wide variety of approaches have failed to achieve disease modification. It has been suggested that the heterogeneity of PD is a major contributing factor to the failure of disease modification trials, and that it is unlikely that a single treatment will be effective in all patients. Precision medicine, using drugs designed to target the pathophysiology in a manner that is specific to each individual with PD, has been suggested as a way forward. PD patients can be stratified according to whether they carry one of the risk variants associated with elevated PD risk. In this review we assess current clinical trials targeting two enzymes, leucine-rich repeat kinase 2 (LRRK2) and glucocerebrosidase (GBA), which are encoded by two most common PD risk genes. Because the details of the pathogenic processes coupled to the different LRRK2 and GBA risk variants are not fully understood, we ask if these precision medicine-based intervention strategies will prove "precise" or "personalized" enough to modify the disease process in PD patients. We also consider at what phases of the disease that such strategies might be effective, in light of the genes being primarily associated with the risk of developing disease in the first place, and less clearly linked to the rate of disease progression. Finally, we critically evaluate the notion that therapies targeting LRRK2 and GBA might be relevant to a wider segment of PD patients, beyond those that actually carry risk variants of these genes.
Collapse
Affiliation(s)
| | - Ziv Gan-Or
- Montreal Neurological Institute, McGill University, Montréal, QC, H3A 2B4, Canada.,Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 2B4, Canada
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
| |
Collapse
|
33
|
Crotty GF, Maciuca R, Macklin EA, Wang J, Montalban M, Davis SS, Alkabsh JI, Bakshi R, Chen X, Ascherio A, Astarita G, Huntwork-Rodriguez S, Schwarzschild MA. Association of caffeine and related analytes with resistance to Parkinson disease among LRRK2 mutation carriers: A metabolomic study. Neurology 2020; 95:e3428-e3437. [PMID: 32999056 PMCID: PMC7836665 DOI: 10.1212/wnl.0000000000010863] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/17/2020] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To identify markers of resistance to developing Parkinson disease (PD) among LRRK2 mutation carriers (LRRK2+), we carried out metabolomic profiling in individuals with PD and unaffected controls (UC), with and without the LRRK2 mutation. METHODS Plasma from 368 patients with PD and UC in the LRRK2 Cohort Consortium (LCC), comprising 118 LRRK2+/PD+, 115 LRRK2+/UC, 70 LRRK2-/PD+, and 65 LRRK2-/UC, and CSF available from 68 of them, were analyzed by liquid chromatography with mass spectrometry. For 282 analytes quantified in plasma and CSF, we assessed differences among the 4 groups and interactions between LRRK2 and PD status, using analysis of covariance models adjusted by age, study site cohort, and sex, with p value corrections for multiple comparisons. RESULTS Plasma caffeine concentration was lower in patients with PD vs UC (p < 0.001), more so among LRRK2+ carriers (by 76%) than among LRRK2- participants (by 31%), with significant interaction between LRRK2 and PD status (p = 0.005). Similar results were found for caffeine metabolites (paraxanthine, theophylline, 1-methylxanthine) and a nonxanthine marker of coffee consumption (trigonelline) in plasma, and in the subset of corresponding CSF samples. Dietary caffeine was also lower in LRRK2+/PD+ compared to LRRK2+/UC with significant interaction effect with the LRRK2+ mutation (p < 0.001). CONCLUSIONS Metabolomic analyses of the LCC samples identified caffeine, its demethylation metabolites, and trigonelline as prominent markers of resistance to PD linked to pathogenic LRRK2 mutations, more so than to idiopathic PD. Because these analytes are known both as correlates of coffee consumption and as neuroprotectants in animal PD models, the findings may reflect their avoidance by those predisposed to develop PD or their protective effects among LRRK2 mutation carriers.
Collapse
Affiliation(s)
- Grace F Crotty
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA.
| | - Romeo Maciuca
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Eric A Macklin
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Junhua Wang
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Manuel Montalban
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Sonnet S Davis
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Jamal I Alkabsh
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Rachit Bakshi
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Xiqun Chen
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Alberto Ascherio
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Giuseppe Astarita
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Sarah Huntwork-Rodriguez
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| | - Michael A Schwarzschild
- From the Department of Neurology (G.F.C., R.B., X.C., M.A.S.) and Biostatistics Center, Department of Medicine (E.A.M.), Massachusetts General Hospital; Harvard Medical School (G.F.C., E.A.M., R.B., X.C., A.A., M.A.S.), Boston, MA; Denali Therapeutics Inc. (R.M., J.W., M.M., S.S.D., J.I.A., G.A., S.H.-R.), San Francisco, CA; and Department of Nutrition (A.A.), Harvard T. H. Chan School of Public Health, Boston, MA
| |
Collapse
|
34
|
Delcambre S, Ghelfi J, Ouzren N, Grandmougin L, Delbrouck C, Seibler P, Wasner K, Aasly JO, Klein C, Trinh J, Pereira SL, Grünewald A. Mitochondrial Mechanisms of LRRK2 G2019S Penetrance. Front Neurol 2020; 11:881. [PMID: 32982917 PMCID: PMC7477385 DOI: 10.3389/fneur.2020.00881] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Several mutations in leucine-rich repeat kinase-2 (LRRK2) have been associated with Parkinson's disease (PD). The most common substitution, G2019S, interferes with LRRK2 kinase activity, which is regulated by autophosphorylation. Yet, the penetrance of this gain-of-function mutation is incomplete, and thus far, few factors have been correlated with disease status in carriers. This includes (i) LRRK2 autophosphorylation in urinary exosomes, (ii) serum levels of the antioxidant urate, and (iii) abundance of mitochondrial DNA (mtDNA) transcription-associated 7S DNA. In light of a mechanistic link between LRRK2 kinase activity and mtDNA lesion formation, we previously investigated mtDNA integrity in fibroblasts from manifesting (LRRK2+/PD+) and non-manifesting carriers (LRRK2+/PD−) of the G2019S mutation as well as from aged-matched controls. In our published study, mtDNA major arc deletions correlated with PD status, with manifesting carriers presenting the highest levels. In keeping with these findings, we now further explored mitochondrial features in fibroblasts derived from LRRK2+/PD+ (n = 10), LRRK2+/PD− (n = 21), and control (n = 10) individuals. In agreement with an accumulation of mtDNA major arc deletions, we also detected reduced NADH dehydrogenase activity in the LRRK2+/PD+ group. Moreover, in affected G2019S carriers, we observed elevated mitochondrial mass and mtDNA copy numbers as well as increased expression of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates antioxidant signaling. Taken together, these results implicate mtDNA dyshomeostasis—possibly as a consequence of impaired mitophagy—in the penetrance of LRRK2-associated PD. Our findings are a step forward in the pursuit of unveiling markers that will allow monitoring of disease progression of LRRK2 mutation carriers.
Collapse
Affiliation(s)
- Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jenny Ghelfi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Nassima Ouzren
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Léa Grandmougin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catherine Delbrouck
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Kobi Wasner
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jan O Aasly
- Department of Neuromedicine and Movement Science, Department of Neurology, St. Olav's Hospital, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Joanne Trinh
- Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Sandro L Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
35
|
Iron-responsive-like elements and neurodegenerative ferroptosis. ACTA ACUST UNITED AC 2020; 27:395-413. [PMID: 32817306 PMCID: PMC7433652 DOI: 10.1101/lm.052282.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022]
Abstract
A set of common-acting iron-responsive 5′untranslated region (5′UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aβ from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem–loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5′UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.
Collapse
|
36
|
Padmanabhan S, Fiske BK, Baptista MA. The Michael J. Fox Foundation's Strategies for Accelerating Translation of LRRK2 into Therapies for Parkinson Disease. Cells 2020; 9:E1878. [PMID: 32796584 PMCID: PMC7466022 DOI: 10.3390/cells9081878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/01/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022] Open
Abstract
Since 2005, The Michael J. Fox Foundation for Parkinson's Research (MJFF) has invested significant funding and non-funding effort to accelerate research and drug development activity around the Parkinson disease (PD)-associated protein LRRK2. MJFF has spearheaded multiple public/private pre-competitive collaborations that have contributed to our understanding of LRRK2 function; de-risked potential safety questions around the therapeutic use of LRRK2 kinase inhibitors; and generated critical research tools, biosamples, and data for the field. Several LRRK2-targeted therapies are now in human testing due to the hard work of so many in the PD community. In this perspective, we present a holistic description and model of how our Foundation's support targeted important barriers to LRRK2 research and helped move the field into clinical trials.
Collapse
Affiliation(s)
- Shalini Padmanabhan
- The Michael J. Fox Foundation for Parkinson’s Research, Grand Central Station, P.O. Box 4777, New York, NY 10120, USA
| | | | - Marco A.S. Baptista
- The Michael J. Fox Foundation for Parkinson’s Research, Grand Central Station, P.O. Box 4777, New York, NY 10120, USA
| |
Collapse
|
37
|
San Luciano M, Tanner CM, Meng C, Marras C, Goldman SM, Lang AE, Tolosa E, Schüle B, Langston JW, Brice A, Corvol JC, Goldwurm S, Klein C, Brockman S, Berg D, Brockmann K, Ferreira JJ, Tazir M, Mellick GD, Sue CM, Hasegawa K, Tan EK, Bressman S, Saunders-Pullman R. Nonsteroidal Anti-inflammatory Use and LRRK2 Parkinson's Disease Penetrance. Mov Disord 2020; 35:1755-1764. [PMID: 32662532 DOI: 10.1002/mds.28189] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/15/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The penetrance of leucine rich repeat kinase 2 (LRRK2) mutations is incomplete and may be influenced by environmental and/or other genetic factors. Nonsteroidal anti-inflammatory drugs (NSAIDs) are known to reduce inflammation and may lower Parkinson's disease (PD) risk, but their role in LRRK2-associated PD is unknown. OBJECTIVES The objective of this study is to evaluate the association of regular NSAID use and LRRK2-associated PD. METHODS Symptomatic ("LRRK2-PD") and asymptomatic ("LRRK2-non-PD") participants with LRRK2 G2019S, R1441X, or I2020T variants (definitely pathogenic variant carriers) or G2385R or R1628P variants (risk variant carriers) from 2 international cohorts provided information on regular ibuprofen and/or aspirin use (≥2 pills/week for ≥6 months) prior to the index date (diagnosis date for PD, interview date for non-PD). Multivariate logistic regression was used to evaluate the relationship between regular NSAID use and PD for any NSAID, separately for ibuprofen and aspirin in all carriers and separately in pathogenic and risk variant groups. RESULTS A total of 259 LRRK2-PD and 318 LRRK2-non-PD participants were enrolled. Regular NSAID use was associated with reduced odds of PD in the overall cohort (odds ratio [OR], 0.34; 95% confidence interval [CI], 0.21-0.57) and in both pathogenic and risk variant carriers (ORPathogenic , 0.38; 95% CI, 0.21-0.67 and ORRiskVariant , 0.19; 95% CI, 0.04-0.99). Similar associations were observed for ibuprofen and aspirin separately (ORIbuprofen , 0.19; 95% CI, 0.07-0.50 and ORAspirin , 0.51; 95% CI, 0.28-0.91). CONCLUSIONS Regular NSAID use may be associated with reduced penetrance in LRRK2-associated PD. The LRRK2 protein is involved in inflammatory pathways and appears to be modulated by regular anti-inflammatory use. Longitudinal observational and interventional studies of NSAID exposure and LRRK2-PD are needed to confirm this association. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marta San Luciano
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Caroline M Tanner
- Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Department of Neurology, San Francisco Veterans Affairs Medical Center, San Francisco, California, USA
| | - Cheryl Meng
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre, Toronto, Ontario, Canada.,Department of Neurology, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Samuel M Goldman
- Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Anthony E Lang
- The Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Centre, Toronto, Ontario, Canada.,Department of Neurology, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Eduardo Tolosa
- Movement Disorders Unit, Neurology Service, Hospital Clínic, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (IDIBAPS) Universitat de Barcelona, Catalonia, Spain
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - J William Langston
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.,Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Alexis Brice
- Sorbonne Universites, UPMC Universite Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Universites, UPMC Universite Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle epiniere, ICM, Paris, France
| | | | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Simone Brockman
- School of Psychiatry and Clinical Neurosciences, University of Western Australia and Fremantle Hospital, Western Australia, Australia
| | - Daniela Berg
- Department for Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Kathrin Brockmann
- Department for Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Joachim J Ferreira
- Clinical Pharmacology Unit, Instituto de Medicina Molecular, Lisbon, Portugal
| | - Meriem Tazir
- Service de Neurologie CHU Mustapha, Alger, Algeria
| | - George D Mellick
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - Carolyn M Sue
- Department of Neurogenetics, Kolling Institute, University of Sydney, Sydney, Australia
| | - Kazuko Hasegawa
- Department of Neurology, Sagamihara National Hospital, Kanagawa, Japan
| | - Eng King Tan
- Department of Neurology, Singapore General Hospital, Singapore
| | - Susan Bressman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | |
Collapse
|
38
|
Loeffler DA, Aasly JO, LeWitt PA, Coffey MP. What Have We Learned from Cerebrospinal Fluid Studies about Biomarkers for Detecting LRRK2 Parkinson's Disease Patients and Healthy Subjects with Parkinson's-Associated LRRK2 Mutations? JOURNAL OF PARKINSONS DISEASE 2020; 9:467-488. [PMID: 31322581 PMCID: PMC6700639 DOI: 10.3233/jpd-191630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common known cause of autosomal dominant Parkinson’s disease (PD) and sporadic PD (sPD). The clinical presentation of LRRK2 PD is similar to sPD, and except for genetic testing, no biochemical or imaging markers can differentiate LRRK2 PD from sPD. Discovery of such biomarkers could indicate neuropathological mechanisms that are unique to or increased in LRRK2 PD. This review discusses findings in 17 LRRK2 - related CSF studies found on PubMed. Most of these studies compared analyte concentrations between four diagnostic groups: LRRK2 PD patients, sPD patients, asymptomatic control subjects carrying PD-associated LRRK2 mutations (LRRK2 CTL), and healthy control subjects lacking LRRK2 mutations (CTL). Analytes examined in these studies included Aβ1-42, tau, α-synuclein, oxidative stress markers, autophagy-related proteins, pteridines, neurotransmitter metabolites, exosomal LRRK2 protein, RNA species, inflammatory cytokines, mitochondrial DNA (mtDNA), and intermediary metabolites. FINDINGS: Pteridines, α-synuclein, mtDNA, 5-hydroxyindolacetic acid, β-D-glucose, lamp2, interleukin-8, and vascular endothelial growth factor were suggested to differentiate LRRK2 PD from sPD patients; 8-hydroxy-2’-deoxyguanosine (8-OHdG), 8-isoprostane (8-ISO), 2-hydroxybutyrate, mtDNA, lamp2, and neopterin may differentiate between LRRK2 CTL and LRRK2 PD subjects; and soluble oligomeric α-synuclein, 8-OHdG, and 8-ISO might differentiate LRRK2 CTL from CTL subjects. CONCLUSIONS: The low numbers of investigations of each analyte, small sample sizes, and methodological differences limit conclusions that can be drawn from these studies. Further investigations are indicated to determine the validity of the analytes identified in these studies as possible biomarkers for LRRK2 PD patients and/or LRRK2 CTL subjects.
Collapse
Affiliation(s)
- David A Loeffler
- Department of Neurology, Beaumont Hospital-Royal Oak, Beaumont Health, Royal Oak, MI, USA
| | - Jan O Aasly
- Department of Neurology, St. Olav's Hospital, Trondheim, Norway
| | - Peter A LeWitt
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mary P Coffey
- Department of Biostatistics, Beaumont Hospital-Royal Oak, Beaumont Health, Royal Oak, MI, USA
| |
Collapse
|
39
|
Cerri S, Mus L, Blandini F. Parkinson's Disease in Women and Men: What's the Difference? JOURNAL OF PARKINSONS DISEASE 2020; 9:501-515. [PMID: 31282427 PMCID: PMC6700650 DOI: 10.3233/jpd-191683] [Citation(s) in RCA: 348] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increasing evidence points to biological sex as an important factor in the development and phenotypical expression of Parkinson’s disease (PD). Risk of developing PD is twice as high in men than women, but women have a higher mortality rate and faster progression of the disease. Moreover, motor and nonmotor symptoms, response to treatments and disease risk factors differ between women and men. Altogether, sex-related differences in PD support the idea that disease development might involve distinct pathogenic mechanisms (or the same mechanism but in a different way) in male and female patients. This review summarizes the most recent knowledge concerning differences between women and men in PD clinical features, risk factors, response to treatments and mechanisms underlying the disease pathophysiology. Unraveling how the pathology differently affect the two sexes might allow the development of tailored interventions and the design of innovative programs that meet the distinct needs of men and women, improving patient care.
Collapse
Affiliation(s)
- Silvia Cerri
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Liudmila Mus
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabio Blandini
- Laboratory of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
40
|
Koros C, Simitsi AM, Papadimitriou D, Bougea A, Prentakis A, Papagiannakis N, Pachi I, Bozi M, Antonelou R, Angelopoulou E, Beratis I, Papageorgiou SG, Trapali XG, Stamelou M, Stefanis L. Serum Uric Acid Level as a Biomarker in Idiopathic and Genetic (p.A53T Alpha-Synuclein Carriers) Parkinson’s Disease: Data from the PPMI Study. JOURNAL OF PARKINSONS DISEASE 2020; 10:481-487. [DOI: 10.3233/jpd-191860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Christos Koros
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
- 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | - Athina-Maria Simitsi
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | | | - Anastasia Bougea
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | - Andreas Prentakis
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
- Nuclear Medicine Unit, Attikon Hospital, Athens, Greece
| | - Nikolaos Papagiannakis
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | - Ioanna Pachi
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
- 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | - Maria Bozi
- 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | - Roubina Antonelou
- 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | - Efthalia Angelopoulou
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| | - Ion Beratis
- 2nd Neurology Clinic, Attikon Hospital, Athens University Medical School, Athens, Greece
| | | | | | - Maria Stamelou
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
- Neurology Clinic, Philipps University, Marburg, Germany
- Parkinson’s disease and Movement Disorders Dept., HYGEIA Hospital, Athens, Greece
| | - Leonidas Stefanis
- 1st Neurology Clinic, Eginition Hospital, Athens University Medical School, Athens, Greece
| |
Collapse
|
41
|
Schirinzi T, Di Lazzaro G, Sancesario GM, Summa S, Petrucci S, Colona VL, Bernardini S, Pierantozzi M, Stefani A, Mercuri NB, Pisani A. Young-onset and late-onset Parkinson's disease exhibit a different profile of fluid biomarkers and clinical features. Neurobiol Aging 2020; 90:119-124. [PMID: 32169356 DOI: 10.1016/j.neurobiolaging.2020.02.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Young-onset Parkinson's disease (YOPD) is a relevant condition whose neurobiology is questioned if different from those of typical late-onset Parkinson's disease (LOPD). Here, we explored whether the clinical-biochemical profile of Parkinson's disease (PD) could be affected by the age-of-onset (AO), as a possible result of a distinct neurodegenerative process. A panel of fluid biomarkers (CSF lactate, 42-amyloid-β peptide, total and 181-phosphorylated tau; serum uric acid) and the standard scores for motor and nonmotor signs were assessed in 76 idiopathic PD patients (genetic cases excluded; YOPD, AO ≤ 50, n = 44; LOPD, AO > 50, n = 32) and 75 sex/age-matched controls, adjusting the models for the main confounding factors. In PD, AO directly correlated to either CSF lactate and tau proteins or the nonmotor symptoms scale score. Specifically, a younger AO was associated with lower levels of biomarkers and minor burden of nonmotor symptoms. Our findings indicate that clinical-biochemical features of idiopathic PD may vary depending on the AO, accounting for different profiles in YOPD and LOPD whose recognition is fundamental for further pathophysiological implications and clinical applications.
Collapse
Affiliation(s)
- Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; Department of Neurosciences, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Giulia Maria Sancesario
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | - Susanna Summa
- Department of Neurosciences, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy; Department of Clinical and Molecular Medicine, S. Andrea University Hospital, Rome, Italy; IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Vito Luigi Colona
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Roma Tor Vergata, Rome, Italy
| | | | - Alessandro Stefani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Antonio Pisani
- Department of Systems Medicine, University of Roma Tor Vergata, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
42
|
Weindel CG, Bell SL, Vail KJ, West KO, Patrick KL, Watson RO. LRRK2 maintains mitochondrial homeostasis and regulates innate immune responses to Mycobacterium tuberculosis. eLife 2020; 9:51071. [PMID: 32057291 PMCID: PMC7159881 DOI: 10.7554/elife.51071] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
The Parkinson’s disease (PD)-associated gene leucine-rich repeat kinase 2 (LRRK2) has been studied extensively in the brain. However, several studies have established that mutations in LRRK2 confer susceptibility to mycobacterial infection, suggesting LRRK2 also controls immunity. We demonstrate that loss of LRRK2 in macrophages induces elevated basal levels of type I interferon (IFN) and interferon stimulated genes (ISGs) and causes blunted interferon responses to mycobacterial pathogens and cytosolic nucleic acid agonists. Altered innate immune gene expression in Lrrk2 knockout (KO) macrophages is driven by a combination of mitochondrial stresses, including oxidative stress from low levels of purine metabolites and DRP1-dependent mitochondrial fragmentation. Together, these defects promote mtDNA leakage into the cytosol and chronic cGAS engagement. While Lrrk2 KO mice can control Mycobacterium tuberculosis (Mtb) replication, they have exacerbated inflammation and lower ISG expression in the lungs. These results demonstrate previously unappreciated consequences of LRRK2-dependent mitochondrial defects in controlling innate immune outcomes. Parkinson’s disease is a progressive nervous system disorder that causes tremors, slow movements, and stiff and inflexible muscles. The symptoms are caused by the loss of cells known as neurons in a specific part of the brain that helps to regulate how the body moves. Researchers have identified mutations in several genes that are associated with an increased risk of developing Parkinson’s. The most common of these mutations occur in a gene called LRRK2. This gene produces a protein that has been shown to be important for maintaining cellular compartments known as mitochondria, which play a crucial role in generating energy. It remains unclear how these mutations lead to the death of neurons. Mutations in LRRK2 have also been shown to make individuals more susceptible to bacterial infections, suggesting that the protein that LRRK2 codes for may help our immune system. Weindel, Bell et al. set out to understand how this protein works in immune cells called macrophages, which ‘eat’ invading bacteria and produce type I interferons, molecules that promote immune responses. Mouse cells were used to measure the ability of normal macrophages and macrophages that lack the mouse equivalent to LRRK2 (referred to as Lrrk2 knockout macrophages) to make type I interferons. The experiments showed that the Lrrk2 knockout macrophages made type I interferons even when they were not infected with bacteria, suggesting they are subject to stress that triggers immune responses. It was possible to correct the behavior of the Lrrk2 knockout macrophages by repairing their mitochondria. When mice missing the gene equivalent to LRRK2 were infected with the bacterium that causes tuberculosis, they experienced more severe disease. The protein encoded by the LRRK2 gene is considered a potential target for therapies to treat Parkinson’s disease, and several drugs that inhibit this protein are being tested in clinical trials. The findings of Weindel, Bell et al. suggest that these drugs may have unintended negative effects on a patient’s ability to fight infection. This work also indicates that LRRK2 mutations may disrupt immune responses in the brain, where macrophage-like cells called microglia play a crucial role in maintaining healthy neurons. Future studies that examine how mutations in LRRK2 affect microglia may help us understand how Parkinson’s disease develops.
Collapse
Affiliation(s)
- Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Krystal J Vail
- Department of Veterinary Pathobiology, Texas A&M University College of Veterinary Medicine and Biomedical Sciences, College Station, United States
| | - Kelsi O West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, United States
| |
Collapse
|
43
|
Bakshi R, Macklin EA, Schwarzschild MA. Reply to “Mitochondrial DNA deletions discriminate affected from unaffected
LRRK
2 mutation carriers”. Ann Neurol 2019; 86:326-327. [DOI: 10.1002/ana.25509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Rachit Bakshi
- Department of NeurologyMassachusetts General Hospital Boston MA
- Harvard Medical School Boston MA
| | - Eric A. Macklin
- Harvard Medical School Boston MA
- Biostatistics Center, Department of MedicineMassachusetts General Hospital Boston MA
| | | |
Collapse
|
44
|
Ouzren N, Delcambre S, Ghelfi J, Seibler P, Farrer MJ, König IR, Aasly JO, Trinh J, Klein C, Grünewald A. Mitochondrial DNA Deletions Discriminate Affected from Unaffected LRRK2 Mutation Carriers. Ann Neurol 2019; 86:324-326. [PMID: 31148195 PMCID: PMC6900150 DOI: 10.1002/ana.25510] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 01/21/2023]
Affiliation(s)
- Nassima Ouzren
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jenny Ghelfi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Matthew J Farrer
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Inke R König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Jan O Aasly
- Department of Neuromedicine and Movement Science and Department of Neurology, St Olav's Hospital, Norwegian University of Science and Technology, Trondheim, Norway
| | - Joanne Trinh
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|