1
|
Shima S, Mizutani Y, Yoshimoto J, Maeda Y, Ohdake R, Nagao R, Maeda T, Higashi A, Ueda A, Ito M, Mutoh T, Watanabe H. Uric acid and alterations of purine recycling disorders in Parkinson's disease: a cross-sectional study. NPJ Parkinsons Dis 2024; 10:170. [PMID: 39251680 PMCID: PMC11385569 DOI: 10.1038/s41531-024-00785-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/20/2024] [Indexed: 09/11/2024] Open
Abstract
The relationship between reduced serum uric acid (UA) levels and Parkinson's disease (PD), particularly purine metabolic pathways, is not fully understood. Our study compared serum and cerebrospinal fluid (CSF) levels of inosine, hypoxanthine, xanthine, and UA in PD patients and healthy controls. We analyzed 132 samples (serum, 45 PD, and 29 age- and sex-matched healthy controls; CSF, 39 PD, and 19 age- and sex-matched healthy controls) using liquid chromatography-tandem mass spectrometry. Results showed significantly lower serum and CSF UA levels in PD patients than in controls (p < 0.0001; effect size r = 0.5007 in serum, p = 0.0046; r = 0.3720 in CSF). Decreased serum hypoxanthine levels were observed (p = 0.0002; r = 0.4338) in PD patients compared to controls with decreased CSF inosine and hypoxanthine levels (p < 0.0001, r = 0.5396: p = 0.0276, r = 0.2893). A general linear model analysis indicated that the reduced UA levels were mainly due to external factors such as sex and weight in serum and age and weight in CSF unrelated to the purine metabolic pathway. Our findings highlight that decreased UA levels in PD are influenced by factors beyond purine metabolism, including external factors such as sex, weight, and age, emphasizing the need for further research into the underlying mechanisms and potential therapeutic approaches.
Collapse
Affiliation(s)
- Sayuri Shima
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuaki Mizutani
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Junichiro Yoshimoto
- Department of Biomedical Data Science, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuhiro Maeda
- Open Facility Center, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Reiko Ohdake
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Ryunosuke Nagao
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Toshiki Maeda
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Atsuhiro Higashi
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Akihiro Ueda
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Mizuki Ito
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Tatsuro Mutoh
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
- Fujita Health University Central Japan International Airport Clinic, 1-1 Centrair, Tokoname, Aichi, 479-0881, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
2
|
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
|
3
|
Zhang YY, Jiang XH, Zhu PP, Zhuo WY, Liu LB. Advancements in understanding substantia nigra hyperechogenicity via transcranial sonography in Parkinson's disease and its clinical implications. Front Neurol 2024; 15:1407860. [PMID: 39091976 PMCID: PMC11291319 DOI: 10.3389/fneur.2024.1407860] [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: 03/27/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Amidst rising Parkinson's disease (PD) incidence in an aging global population, the need for non-invasive and reliable diagnostic methods is increasingly critical. This review evaluates the strategic role of transcranial sonography (TCS) in the early detection and monitoring of PD. TCS's ability to detect substantia nigra hyperechogenicity offers profound insights into its correlation with essential neuropathological alterations-namely, iron accumulation, neuromelanin depletion, and glial proliferation-fundamental to PD's pathophysiology. Our analysis highlights TCS's advantages, including its non-invasiveness, cost-effectiveness, and ease of use, positioning it as an invaluable tool for early diagnosis and continual disease progression monitoring. Moreover, TCS assists in identifying potential risk and protective factors, facilitating tailored therapeutic strategies to enhance clinical outcomes. This review advocates expanding TCS utilization and further research to maximize its diagnostic and prognostic potential in PD management, contributing to a more nuanced understanding of the disease.
Collapse
Affiliation(s)
- Yuan-yuan Zhang
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Xu-hong Jiang
- Department of Health Management, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Pei-pei Zhu
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Wen-yan Zhuo
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| | - Li-bin Liu
- Department of Neurology, Zhuhai People’s Hospital, Zhuhai, Guangdong, China
| |
Collapse
|
4
|
Zhang T, Lv H, Zhao X, Zeng N, Hui Y, Chen S, Wu N, Xu M, Wu Y, Xing A, Shi H, Zhang S, Liang X, Wang Y, Wu S, Cui L, Wang Z, Liu Y. Serum uric acid is associated with midbrain enlarged perivascular spaces: Results from Multi-modality Medical imaging sTudy bAsed on KaiLuan Study (META-KLS). Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111000. [PMID: 38580007 DOI: 10.1016/j.pnpbp.2024.111000] [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: 12/07/2023] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Serum uric acid (SUA) is a major cause of cardiovascular and cerebrovascular diseases. Whether and to what extent the excess risk of enlarged perivascular spaces (EPVS) conferred by SUA is unknown. The study was conducted to investigate the association between SUA and EPVS in different brain regions. METHODS Data are from Multi-modality medical imaging study based on Kailuan study (META-KLS) in this cross-sectional study. Participants were divided into five groups based on SUA levels, and EPVS in basal ganglia (BG), centrum semiovale (CSO) and midbrain (MB) was systematically assessed and divided into Low and High group. Odds ratio (OR) and 95% confidence intervals (95% CIs) for high EPVS outcomes were estimated using multivariable logistic regression analysis. Restricted cubic spline (RCS) was used to further investigate dose-response relationship. RESULTS A total of 1014 participants aged 25-83 years from 11 centers were enrolled in the study. In the multivariable-adjusted model, SUA, as an independent risk factor, correlated positively with high degree of MB-EPVS (OR, 1.002; 95% CI, 1.000 to 1.004; p = 0.023) in general population. In addition, RCS further demonstrated the linear association between SUA and MB-EPVS (p = 0.072). No association was found between SUA and BG-EPVS or CSO-EPVS. CONCLUSION SUA was an independent risk factor of MB-EPVS. High SUA levels were more predictive of increased risk occurrence of high degree of MB-EPVS, supporting a linear association between SUA and MB-EPVS and further indicating that SUA may play an important role in cerebral small vessel disease. TRIAL REGISTRATION The KaiLuan Study and META-KLS were registered online (ChiCTR2000029767 on chictr.org.cn and NCT05453877 on Clinicaltrials.gov, respectively).
Collapse
Affiliation(s)
- Tong Zhang
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Han Lv
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinyu Zhao
- Clinical Epidemiology & EBM Unit, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Na Zeng
- School of Public Health, Peking University, Beijing, China
| | - Ying Hui
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shuohua Chen
- Department of Cardiovascular Lab, Kailuan General Hospital, Tangshan, Hebei, China
| | - Ning Wu
- Department of Medical Imaging, Yanjing Medical College, Capital Medical University, Beijing, China
| | - Mingze Xu
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Beijing Intelligent Brain Cloud Inc, Beijing, China
| | - YunTao Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Aijun Xing
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Huijing Shi
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Shun Zhang
- Department of Psychiatry, Kailuan Mental Health Center, Tangshan, Hebei, China
| | - Xiaoliang Liang
- Department of Psychiatry, Kailuan Mental Health Center, Tangshan, Hebei, China
| | - Yongxin Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of MR, Kailuan General Hospital, Tangshan, Hebei, China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Liufu Cui
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yanying Liu
- Department of Rheumatology and Immunology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Sodders MJ, Avila-Pacheco J, Okorie EC, Shen M, Kumari N, Marathi A, Lakhani M, Bullock K, Pierce K, Dennis C, Jeanfavre S, Sarkar S, Scherzer CR, Clish C, Olsen AL. Genetic screening and metabolomics identify glial adenosine metabolism as a therapeutic target in Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594309. [PMID: 38798570 PMCID: PMC11118494 DOI: 10.1101/2024.05.15.594309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and lacks disease-modifying therapies. We developed a Drosophila model for identifying novel glial-based therapeutic targets for PD. Human alpha-synuclein is expressed in neurons and individual genes are independently knocked down in glia. We performed a forward genetic screen, knocking down the entire Drosophila kinome in glia in alpha-synuclein expressing flies. Among the top hits were five genes (Ak1, Ak6, Adk1, Adk2, and awd) involved in adenosine metabolism. Knockdown of each gene improved locomotor dysfunction, rescued neurodegeneration, and increased brain adenosine levels. We determined that the mechanism of neuroprotection involves adenosine itself, as opposed to a downstream metabolite. We dove deeper into the mechanism for one gene, Ak1, finding rescue of dopaminergic neuron loss, alpha-synuclein aggregation, and bioenergetic dysfunction after glial Ak1 knockdown. We performed metabolomics in Drosophila and in human PD patients, allowing us to comprehensively characterize changes in purine metabolism and identify potential biomarkers of dysfunctional adenosine metabolism in people. These experiments support glial adenosine as a novel therapeutic target in PD.
Collapse
|
6
|
Koh S, Lee DY, Cha JM, Kim Y, Kim HH, Yang HJ, Park RW, Choi JY. Association between pre-diagnostic serum uric acid levels in patients with newly diagnosed epilepsy and conversion rate to drug-resistant epilepsy within 5 years: A common data model analysis. Seizure 2024; 118:103-109. [PMID: 38669746 DOI: 10.1016/j.seizure.2024.04.014] [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: 01/16/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
PURPOSE Drug-resistant epilepsy (DRE) poses a significant challenge in epilepsy management, and reliable biomarkers for identifying patients at risk of DRE are lacking. This study aimed to investigate the association between serum uric acid (UA) levels and the conversion rate to DRE. METHODS A retrospective cohort study was conducted using a common data model database. The study included patients newly diagnosed with epilepsy, with prediagnostic serum UA levels within a six-month window. Patients were categorized into hyperUA (≥7.0 mg/dL), normoUA (<7.0 and >2.0 mg/dL), and hypoUA (≤2.0 mg/dL) groups based on their prediagnostic UA levels. The outcome was the conversion rate to DRE within five years of epilepsy diagnosis. RESULTS The study included 5,672 patients with epilepsy and overall conversion rate to DRE was 19.4%. The hyperUA group had a lower DRE conversion rate compared to the normoUA group (HR: 0.81 [95% CI: 0.69-0.96]), while the hypoUA group had a higher conversion rate (HR: 1.88 [95% CI: 1.38-2.55]). CONCLUSIONS Serum UA levels have the potential to serve as a biomarker for identifying patients at risk of DRE, indicating a potential avenue for novel therapeutic strategies aimed at preventing DRE conversion.
Collapse
Affiliation(s)
- Seungyon Koh
- Department of Brain Science, Ajou University School of Medicine, 164 World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea; Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Yun Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Kore; Department of Medical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea; Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jae Myung Cha
- Department of Gastroenterology, Gang Dong Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Yerim Kim
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Hyung Hoi Kim
- Department of Laboratory Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Hyeon-Jong Yang
- Department of Pediatrics, Soonchunhyang University College of Medicine, Seoul, Republic of Korea
| | - Rae Woong Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Kore.
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine, 164 World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, Suwon 16499, Republic of Korea; Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
7
|
Yuan X, Wan L, Chen Z, Long Z, Chen D, Liu P, Fu Y, Zhu S, Peng L, Qiu R, Tang B, Jiang H. Peripheral Inflammatory and Immune Landscape in Multiple System Atrophy: A Cross-Sectional Study. Mov Disord 2024; 39:391-399. [PMID: 38155513 DOI: 10.1002/mds.29674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/16/2023] [Accepted: 11/09/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Neuroinflammation might contribute to the pathogenesis of multiple systemic atrophy (MSA). However, specific alterations in the peripheral inflammatory and immune profiles of patients with MSA remain unclear. OBJECTIVES To determine the peripheral inflammatory and immune profiles of patients with MSA and their potential value as biomarkers for facilitating clinical diagnosis and monitoring disease severity. METHODS This cross-sectional study included 235, 240, and 235 patients with MSA, patients with Parkinson's disease (PD), and healthy controls (HCs), respectively. Inflammatory and immune parameters were measured in peripheral blood, differences between groups were assessed, and clusters were analyzed. Associations between the parameters and clinical characteristics of MSA were assessed using Spearman and partial correlation analyses. RESULTS Significant differences were observed especially in monocytes, neutrophils-to-lymphocyte ratio (NLR) and neutrophils-to-lymphocyte ratio (MPV) between MSA patients and HCs (P < 0.01). Monocytes and uric acid (UA) levels were also significantly different between the MSA and PD patients (P < 0.05). The combination of NLR and MPV distinguished MSA-P patients from HCs (areas under the curve = 0.824). In addition, complement components C4 and C3 were significantly correlated with the Scale Outcomes in PD for Autonomic Symptoms and Wexner scale, whereas immunoglobulin G (IgG) was significantly correlated with scores of Unified Multiple System Atrophy Rating Scale (P < 0.05). CONCLUSIONS In MSA patients, monocytes, NLR and MPV might serve as potential diagnostic biomarkers, whereas MLR, C3, C4, and IgG significantly correlate with disease severity. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xinrong Yuan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Linlin Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Zhe Long
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Daji Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Panyan Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - You Fu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Sudan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Linliu Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Qiu
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- National International Collaborative Research Center for Medical Metabolomics, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
8
|
Christine CW, Auinger P, Forti EA, Tat L, Cannizzaro N, Mustafa A, Iyer JM, Oakes D, Green R. Relationships of B12 and Homocysteine with Outcomes in the SURE-PD, SURE-PD3, and STEADY-PDIII Trials. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1243-1255. [PMID: 39240650 PMCID: PMC11380300 DOI: 10.3233/jpd-240035] [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: 06/04/2024] [Indexed: 09/07/2024]
Abstract
Background DATATOP was a study of early Parkinson's disease (PD) conducted in the 1980 s, before mandatory folic acid fortification in the United States. Our analysis of its baseline serum samples revealed a geometric mean vitamin B12 of 369 pg/mL and homocysteine (tHcy) of 9.5μmol/l. We also found that low B12 predicted greater worsening of ambulatory capacity (AC) and elevated tHcy (>15μmol/L) predicted greater declines in cognitive function. Objective We sought to measure B12 and tHcy in contemporary trial participants with early PD who had not started dopaminergic treatment and to determine whether these analytes were associated with clinical progression. Methods We measured B12 and tHcy from baseline and end-of-study blood samples from three recent clinical trials. Results Baseline geometric mean B12 levels for these studies ranged from 484- 618 pg/ml and for tHcy ranged from 7.4- 10μmol/L. Use of B12-containing supplements ranged from 41- 61%, and those taking supplements had higher B12 and lower tHcy. Those who began levodopa, but were not taking B12-supplements, had greater end-of-study tHcy. There was no association of baseline tHcy > 15μmol/L with annualized change in Montreal Cognitive Assessment and no association of baseline B12 tertiles with change in AC. Conclusions In these longitudinal trials, B12 levels were higher than for DATATOP, due in large part to increased B12-supplement intake, while tHcy levels were similar. Initiation of levodopa was associated with increases of tHcy in those not taking a B12-containing supplement. These smaller studies did not replicate prior findings of low B12 and elevated tHcy with features of progression, possibly due to higher baseline B12.
Collapse
Affiliation(s)
- Chadwick W. Christine
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Peggy Auinger
- Department of Neurology, Center for Health and Technology, University of Rochester, Rochester, NY, USA
| | - Esther A.R. Forti
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, CA, USA
| | - Lyvin Tat
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, CA, USA
| | - Noemi Cannizzaro
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, CA, USA
| | - Arshi Mustafa
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, CA, USA
| | - Jay M. Iyer
- Departments of Molecular and Cellular Biology and Statistics, Harvard University, Cambridge, MA, USA
| | - David Oakes
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
9
|
Pfeifer A, Mikhael M, Niemann B. Inosine: novel activator of brown adipose tissue and energy homeostasis. Trends Cell Biol 2024; 34:72-82. [PMID: 37188562 DOI: 10.1016/j.tcb.2023.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023]
Abstract
Extracellular purinergic molecules act as signaling molecules that bind to cellular receptors and regulate signaling pathways. Growing evidence suggests that purines regulate adipocyte function and whole-body metabolism. Here, we focus on one specific purine: inosine. Brown adipocytes, which are important regulators of whole-body energy expenditure (EE), release inosine when they are stressed or become apoptotic. Unexpectedly, inosine activates EE in neighboring brown adipocytes and enhances differentiation of brown preadipocytes. Increasing extracellular inosine, either directly by increasing inosine intake or indirectly via pharmacological inhibition of cellular inosine transporters, increases whole-body EE and counteracts obesity. Thus, inosine and other closely related purines might be a novel approach to tackle obesity and associated metabolic disorders by enhancing EE.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| | - Mickel Mikhael
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Birte Niemann
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| |
Collapse
|
10
|
Ziqubu K, Dludla PV, Mabhida SE, Jack BU, Keipert S, Jastroch M, Mazibuko-Mbeje SE. Brown adipose tissue-derived metabolites and their role in regulating metabolism. Metabolism 2024; 150:155709. [PMID: 37866810 DOI: 10.1016/j.metabol.2023.155709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/28/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
The discovery and rejuvenation of metabolically active brown adipose tissue (BAT) in adult humans have offered a new approach to treat obesity and metabolic diseases. Beyond its accomplished role in adaptive thermogenesis, BAT secretes signaling molecules known as "batokines", which are instrumental in regulating whole-body metabolism via autocrine, paracrine, and endocrine action. In addition to the intrinsic BAT metabolite-oxidizing activity, the endocrine functions of these molecules may help to explain the association between BAT activity and a healthy systemic metabolic profile. Herein, we review the evidence that underscores the significance of BAT-derived metabolites, especially highlighting their role in controlling physiological and metabolic processes involving thermogenesis, substrate metabolism, and other essential biological processes. The conversation extends to their capacity to enhance energy expenditure and mitigate features of obesity and its related metabolic complications. Thus, metabolites derived from BAT may provide new avenues for the discovery of metabolic health-promoting drugs with far-reaching impacts. This review aims to dissect the complexities of the secretory role of BAT in modulating local and systemic metabolism in metabolic health and disease.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho 2745, South Africa
| | - Phiwayinkosi V Dludla
- Cochrane South Africa, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Sihle E Mabhida
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Babalwa U Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Susanne Keipert
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | | |
Collapse
|
11
|
Ciocca M, Pizzamiglio C. Clinical Benefits of Therapeutic Interventions Targeting Mitochondria in Parkinson's Disease Patients. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:554-561. [PMID: 37005519 PMCID: PMC11071650 DOI: 10.2174/1871527322666230330122444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 04/04/2023]
Abstract
Parkinson's disease is the second most common neurodegenerative disease. Mitochondrial dysfunction has been associated with neurodegeneration in Parkinson's disease, and several treatments targeting mitochondria have been tested in these patients to delay disease progression and tackle disease symptoms. Herein, we review available data from randomised, double-blind clinical studies that have investigated the role of compounds targeting mitochondria in idiopathic Parkinson's disease patients, with a view of providing patients and clinicians with a comprehensive and practical paper that can inform therapeutic interventions in this group of people. A total of 9 compounds have been tested in randomized clinical trials, but only exenatide has shown some promising neuroprotective and symptomatic effects. However, whether this evidence can be translated into daily clinical practice still needs to be confirmed. In conclusion, targeting mitochondrial dysfunction in Parkinson's disease is a promising therapeutic approach, although only one compound has shown a positive effect on Parkinson's disease progression and symptoms. New compounds have been investigated in animal models, and their efficacy needs to be confirmed in humans through robust, randomised, double-blind clinical trials.
Collapse
Affiliation(s)
- Matteo Ciocca
- Department of Brain Sciences, Imperial College London, London, UK
| | - Chiara Pizzamiglio
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
12
|
Paul KC, Zhang K, Walker DI, Sinsheimer J, Yu Y, Kusters C, Del Rosario I, Folle AD, Keener AM, Bronstein J, Jones DP, Ritz B. Untargeted serum metabolomics reveals novel metabolite associations and disruptions in amino acid and lipid metabolism in Parkinson's disease. Mol Neurodegener 2023; 18:100. [PMID: 38115046 PMCID: PMC10731845 DOI: 10.1186/s13024-023-00694-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Untargeted high-resolution metabolomic profiling provides simultaneous measurement of thousands of metabolites. Metabolic networks based on these data can help uncover disease-related perturbations across interconnected pathways. OBJECTIVE Identify metabolic disturbances associated with Parkinson's disease (PD) in two population-based studies using untargeted metabolomics. METHODS We performed a metabolome-wide association study (MWAS) of PD using serum-based untargeted metabolomics data derived from liquid chromatography with high-resolution mass spectrometry (LC-HRMS) using two distinct population-based case-control populations. We also combined our results with a previous publication of 34 metabolites linked to PD in a large-scale, untargeted MWAS to assess external validation. RESULTS LC-HRMS detected 4,762 metabolites for analysis (HILIC: 2716 metabolites; C18: 2046 metabolites). We identified 296 features associated with PD at FDR<0.05, 134 having a log2 fold change (FC) beyond ±0.5 (228 beyond ±0.25). Of these, 104 were independently associated with PD in both discovery and replication studies at p<0.05 (170 at p<0.10), while 27 were associated with levodopa-equivalent dose among the PD patients. Intriguingly, among the externally validated features were the microbial-related metabolites, p-cresol glucuronide (FC=2.52, 95% CI=1.67, 3.81, FDR=7.8e-04) and p-cresol sulfate. P-cresol glucuronide was also associated with motor symptoms among patients. Additional externally validated metabolites associated with PD include phenylacetyl-L-glutamine, trigonelline, kynurenine, biliverdin, and pantothenic acid. Novel associations include the anti-inflammatory metabolite itaconate (FC=0.79, 95% CI=0.73, 0.86; FDR=2.17E-06) and cysteine-S-sulfate (FC=1.56, 95% CI=1.39, 1.75; FDR=3.43E-11). Seventeen pathways were enriched, including several related to amino acid and lipid metabolism. CONCLUSIONS Our results revealed PD-associated metabolites, confirming several previous observations, including for p-cresol glucuronide, and newly implicating interesting metabolites, such as itaconate. Our data also suggests metabolic disturbances in amino acid and lipid metabolism and inflammatory processes in PD.
Collapse
Affiliation(s)
- Kimberly C Paul
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Keren Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Janet Sinsheimer
- Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Yu Yu
- Center for Health Policy Research, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Cynthia Kusters
- Department of Human Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Irish Del Rosario
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Aline Duarte Folle
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Adrienne M Keener
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
- Parkinson's Disease Research, Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA, USA
| | - Jeff Bronstein
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Beate Ritz
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| |
Collapse
|
13
|
Zeissler ML, Boey T, Chapman D, Rafaloff G, Dominey T, Raphael KG, Buff S, Pai HV, King E, Sharpe P, O'Brien F, Carroll CB. Investigating trial design variability in trials of disease-modifying therapies in Parkinson's disease: a scoping review protocol. BMJ Open 2023; 13:e071641. [PMID: 38070893 PMCID: PMC10729184 DOI: 10.1136/bmjopen-2023-071641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 11/05/2023] [Indexed: 12/18/2023] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is a debilitating neurological disorder for which the identification of disease-modifying interventions represents a major unmet need. Diverse trial designs have attempted to mitigate challenges of population heterogeneity, efficacious symptomatic therapy and lack of outcome measures that are objective and sensitive to change in a disease modification setting. It is not clear whether consensus is emerging regarding trial design choices. Here, we report the protocol of a scoping review that will provide a contemporary update on trial design variability for disease-modifying interventions in PD. METHODS AND ANALYSIS The Population, Intervention, Comparator, Outcome and Study design (PICOS) framework will be used to structure the review, inform study selection and analysis. The databases MEDLINE, Web of Science, Cochrane and the trial registry ClinicalTrials.gov will be systematically searched to identify published studies and registry entries in English. Two independent reviewers will screen study titles, abstracts and full text for eligibility, with disagreements being resolved through discussion or by a third reviewer where necessary. Data on general study information, eligibility criteria, outcome measures, trial design, retention and statistically significant findings will be extracted into a standardised form. Extracted data will be presented in a descriptive analysis. We will report our findings using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Scoping Review extension. ETHICS AND DISSEMINATION This work will provide an overview of variation and emerging trends in trial design choices for disease-modifying trials of PD. Due to the nature of this study, there are no ethical or safety considerations. We plan to publish our findings in a peer-reviewed journal.
Collapse
Affiliation(s)
- Marie-Louise Zeissler
- Newcastle University, Newcastle upon Tyne, UK
- Faculty of Health, University of Plymouth, Plymouth, UK
| | - Timothy Boey
- School of Medicine, University of Liverpool, Liverpool, Merseyside, UK
| | - Danny Chapman
- Faculty of Health, University of Plymouth, Plymouth, UK
| | - Gary Rafaloff
- Parkinson's Research Advocate, Westlake, Florida, USA
| | - Thea Dominey
- Faculty of Health, University of Plymouth, Plymouth, UK
| | - Karen G Raphael
- Oral & Maxillofacial, Radiology and Medicine, New York University, Brooklyn, New York, USA
- Parkinson's Research Advocate, New York, New York, USA
| | - Susan Buff
- Parkinson's Research Advocate, Sunnyvale, California, USA
| | | | - Emma King
- University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Paul Sharpe
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Camille B Carroll
- Newcastle University, Newcastle upon Tyne, UK
- Faculty of Health, University of Plymouth, Plymouth, UK
| |
Collapse
|
14
|
Henrich MT, Oertel WH, Surmeier DJ, Geibl FF. Mitochondrial dysfunction in Parkinson's disease - a key disease hallmark with therapeutic potential. Mol Neurodegener 2023; 18:83. [PMID: 37951933 PMCID: PMC10640762 DOI: 10.1186/s13024-023-00676-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023] Open
Abstract
Mitochondrial dysfunction is strongly implicated in the etiology of idiopathic and genetic Parkinson's disease (PD). However, strategies aimed at ameliorating mitochondrial dysfunction, including antioxidants, antidiabetic drugs, and iron chelators, have failed in disease-modification clinical trials. In this review, we summarize the cellular determinants of mitochondrial dysfunction, including impairment of electron transport chain complex 1, increased oxidative stress, disturbed mitochondrial quality control mechanisms, and cellular bioenergetic deficiency. In addition, we outline mitochondrial pathways to neurodegeneration in the current context of PD pathogenesis, and review past and current treatment strategies in an attempt to better understand why translational efforts thus far have been unsuccessful.
Collapse
Affiliation(s)
- Martin T Henrich
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Fanni F Geibl
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany.
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany.
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
15
|
Roman YM. The Role of Uric Acid in Human Health: Insights from the Uricase Gene. J Pers Med 2023; 13:1409. [PMID: 37763176 PMCID: PMC10532990 DOI: 10.3390/jpm13091409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Uric acid is the final product of purine metabolism and is converted to allantoin in most mammals via the uricase enzyme. The accumulation of loss of function mutations in the uricase gene rendered hominoids (apes and humans) to have higher urate concentrations compared to other mammals. The loss of human uricase activity may have allowed humans to survive environmental stressors, evolution bottlenecks, and life-threatening pathogens. While high urate levels may contribute to developing gout and cardiometabolic disorders such as hypertension and insulin resistance, low urate levels may increase the risk for neurodegenerative diseases. The double-edged sword effect of uric acid has resurrected a growing interest in urate's antioxidant role and the uricase enzyme's role in modulating the risk of obesity. Characterizing both the effect of uric acid levels and the uricase enzyme in different animal models may provide new insights into the potential therapeutic benefits of uric acid and novel uricase-based therapy.
Collapse
Affiliation(s)
- Youssef M Roman
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
16
|
Arshad U, Rahman F, Hanan N, Chen C. Longitudinal Meta-Analysis of Historical Parkinson's Disease Trials to Inform Future Trial Design. Mov Disord 2023; 38:1716-1727. [PMID: 37400277 DOI: 10.1002/mds.29514] [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: 01/16/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND The outcome of clinical trials in neurodegeneration can be highly uncertain due to the presence of a strong placebo effect. OBJECTIVES To develop a longitudinal model that can enhance the success of future Parkinson's disease trials by quantifying trial-to-trial variations in placebo and active treatment response. METHODS A longitudinal model-based meta-analysis was conducted on the total score of Unified Parkinson's Disease Rating Scale (UPDRS) Parts 1, 2, and 3. The analysis included aggregate data from 66 arms (observational [4], placebo [28], or investigational-drug-treated [34]) from 4 observational studies and 17 interventional trials. Inter-study variabilities in key parameters were estimated. Residual variability was weighted by the size of study arms. RESULTS The baseline total UPDRS was estimated to average at 24.5 points. Disease score was estimated to worsen by 3.90 points/year for the duration of the treatments; whilst notably, arms with a lower baseline progressed faster. The model captured the transient nature of the placebo response and sustained symptomatic drug effect. Both placebo and drug effects peaked within 2 months; although, 1 year was needed to observe the full treatment difference. Across these studies, the progression rate varied by 59.4%, the half-life for offset of placebo response varied by 79.4%, and the amplitude for drug effect varied by 105.3%. CONCLUSION The longitudinal model-based meta-analysis describes UPDRS progression rate, captures the dynamics of the placebo response, quantifies the effect size of the available therapies, and sets the expectation of uncertainty for future trials. The findings provide informative priors to enhance the rigor and success of future trials of promising agents, including potential disease modifiers. © 2023 GSK. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Usman Arshad
- Clinical Pharmacology Modeling and Simulation, GSK, Upper Providence, Pennsylvania, USA
| | - Fatima Rahman
- Clinical Pharmacology Modeling and Simulation, GSK, Upper Providence, Pennsylvania, USA
| | - Nathan Hanan
- Clinical Pharmacology Modeling and Simulation, GSK, Upper Providence, Pennsylvania, USA
| | - Chao Chen
- Clinical Pharmacology Modeling and Simulation, GSK, Upper Providence, Pennsylvania, USA
| |
Collapse
|
17
|
El-Latif AMA, Rabie MA, Sayed RH, Fattah MAAE, Kenawy SA. Inosine attenuates rotenone-induced Parkinson's disease in rats by alleviating the imbalance between autophagy and apoptosis. Drug Dev Res 2023; 84:1159-1174. [PMID: 37170799 DOI: 10.1002/ddr.22077] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 05/13/2023]
Abstract
Growing evidence points to impaired autophagy as one of the major factors implicated in the pathophysiology of Parkinson's disease (PD). Autophagy is a downstream target of adenosine monophosphate-activated protein kinase (AMPK). Inosine has already demonstrated a neuroprotective effect against neuronal loss in neurodegenerative diseases, mainly due its anti-inflammatory and antioxidant properties. We, herein, aimed at investigating the neuroprotective effects of inosine against rotenone-induced PD in rats and to focus on the activation of AMPK-mediated autophagy. Inosine successfully increased p-AMPK/AMPK ratio in PD rats and improved their motor performance and muscular co-ordination (assessed by rotarod, open field, and grip strength tests, as well as by manual gait analysis). Furthermore, inosine was able to mitigate the rotenone-induced histopathological alterations and to restore the tyrosine hydroxylase immunoreactivity in PD rats' substantia nigra. Inosine-induced AMPK activation resulted in an autophagy enhancement, as demonstrated by the increased striatal Unc-S1-like kinase1 and beclin-1 expression, and also by the increment light chain 3II to light chain 3I ratio, along with the decline in striatal mammalian target of rapamycin and p62 protein expressions. The inosine-induced stimulation of AMPK also attenuated neuronal apoptosis and promoted antioxidant activity. Unsurprisingly, these neuroprotective effects were antagonized by a preadministration of dorsomorphin (an AMPK inhibitor). In conclusion, inosine exerted neuroprotective effects against the rotenone-induced neuronal loss via an AMPK activation and through the restoration of the imbalance between autophagy and apoptosis. These findings support potential application of inosine in PD treatment.
Collapse
Affiliation(s)
- Aya M Abd El-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mai A Abd El Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sanaa A Kenawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
18
|
Khanal S, Bok E, Kim J, Park GH, Choi DY. Dopaminergic neuroprotective effects of inosine in MPTP-induced parkinsonian mice via brain-derived neurotrophic factor upregulation. Neuropharmacology 2023:109652. [PMID: 37422180 DOI: 10.1016/j.neuropharm.2023.109652] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. However, no curative or modifying therapy is known. Inosine is a purine nucleoside that increases brain-derived neurotrophic factor (BDNF) expression in the brain through adenosine receptors. Herein, we investigated the neuroprotective effects of inosine and elucidated the mechanisms underlying its pharmacological action. Inosine rescued SH-SY5Y neuroblastoma cells from MPP+ injury in a dose-dependent manner. Inosine protection correlated with BDNF expression and the activation of its downstream signaling cascade, as the TrkB receptor inhibitor, K252a and siRNA against the BDNF gene remarkably reduced the protective effects of inosine. Blocking the A1 or A2A adenosine receptors diminished BDNF induction and the rescuing effect of inosine, indicating a critical role of adenosine A1 and A2A receptors in inosine-related BDNF elevation. We assessed whether the compound could protect dopaminergic neurons from MPTP-induced neuronal injury. Beam-walking and challenge beam tests revealed that inosine pretreatment for 3 weeks reduced the MPTP-induced motor function impairment. Inosine ameliorated dopaminergic neuronal loss and MPTP-mediated astrocytic and microglial activation in the substantia nigra and striatum. Inosine ameliorated the depletion of striatal dopamine and its metabolite following MPTP injection. BDNF upregulation and the activation of its downstream signaling pathway seemingly correlate with the neuroprotective effects of inosine. To our knowledge, this is the first study to demonstrate the neuroprotective effects of inosine against MPTP neurotoxicity via BDNF upregulation. These findings highlight the therapeutic potential of inosine in dopaminergic neurodegeneration in PD brains.
Collapse
Affiliation(s)
- Shristi Khanal
- College of Pharmacy, Yeungnam University, 280 Daehakro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| | - Eugene Bok
- Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| | - Gyu Hwan Park
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280 Daehakro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| |
Collapse
|
19
|
Ong WY, Leow DMK, Herr DR, Yeo CJJ. What Do Randomized Controlled Trials Inform Us About Potential Disease-Modifying Strategies for Parkinson's Disease? Neuromolecular Med 2023; 25:1-13. [PMID: 35776238 DOI: 10.1007/s12017-022-08718-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/05/2022] [Indexed: 01/09/2023]
Abstract
Research advances have shed new insight into cellular pathways contributing to PD pathogenesis and offer increasingly compelling therapeutic targets. In this review, we made a broad survey of the published literature that report possible disease-modifying effects on PD. While there are many studies that demonstrate benefits for various therapies for PD in animal and human studies, we confined our search to human "randomised controlled trials" and with the key words "neuroprotection" or "disease-modifying". It is hoped that through studying the results of these trials, we might clarify possible mechanisms that underlie idiopathic PD. This contrasts with studying the effect of pathophysiology of familial PD, which could be carried out by gene knockouts and animal models. Randomised controlled trials indicate promising effects of MAO-B inhibitors, dopamine agonists, NMDA receptor antagonists, metabotropic glutamate receptor antagonists, therapies related to improving glucose utilization and energy production, therapies related to reduction of excitotoxicity and oxidative stress, statin use, therapies related to iron chelation, therapies related to the use of phytochemicals, and therapies related to physical exercise and brain reward pathway on slowing PD progression. Cumulatively, these approaches fall into two categories: direct enhancement of dopaminergic signalling, and reduction of neurodegeneration. Overlaps between the two categories result in challenges in distinguishing between symptomatic versus disease-modifying effects with current clinical trial designs. Nevertheless, a broad-based approach allows us to consider all possible therapeutic avenues which may be neuroprotective. While the traditional standard of care focuses on symptomatic management with dopaminergic drugs, more recent approaches suggest ways to preserve dopaminergic neurons by attenuating excitotoxicity and oxidative stress.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore.
| | - Damien Meng-Kiat Leow
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Crystal Jing-Jing Yeo
- Institute of Molecular and Cell Biology, A*Star, Singapore, 138673, Singapore
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
- LKC School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- National Neuroscience Institute, Singapore, 308433, Singapore
| |
Collapse
|
20
|
Walk D, Nicholson K, Locatelli E, Chan J, Macklin EA, Ferment V, Manousakis G, Chase M, Connolly M, Dagostino D, Hall M, Ostrow J, Pothier L, Lieberman C, Gelevski D, Randall R, Sherman AV, Steinhart E, Walker DG, Walker J, Yu H, Wills AM, Schwarzschild MA, Beukenhorst AL, Onnela JP, Berry JD, Cudkowicz ME, Paganoni S. Randomized trial of inosine for urate elevation in amyotrophic lateral sclerosis. Muscle Nerve 2023; 67:378-386. [PMID: 36840949 DOI: 10.1002/mus.27807] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 02/26/2023]
Abstract
INTRODUCTION/AIMS Higher urate levels are associated with improved ALS survival in retrospective studies, however whether raising urate levels confers a survival advantage is unknown. In the Safety of Urate Elevation in Amyotrophic Lateral Sclerosis (SURE-ALS) trial, inosine raised serum urate and was safe and well-tolerated. The SURE-ALS2 trial was designed to assess longer term safety. Functional outcomes and a smartphone application were also explored. METHODS Participants were randomized 2:1 to inosine (n = 14) or placebo (n = 9) for 20 weeks, titrated to serum urate of 7-8 mg/dL. Primary outcomes were safety and tolerability. Functional outcomes were measured with the Amyotrophic Lateral Sclerosis Functional Rating Scale Revised (ALSFRS-R). Mobility and ALSFRS-R were also assessed by a smartphone application. RESULTS During inosine treatment, mean urate ranged 5.68-6.82 mg/dL. Treatment-emergent adverse event (TEAE) incidence was similar between groups (p > .10). Renal TEAEs occurred in three (21%) and hypertension in one (7%) of participants randomized to inosine. Inosine was tolerated in 71% of participants versus placebo 67%. Two participants (14%) in the inosine group experienced TEAEs deemed related to treatment (nephrolithiasis); one was a severe adverse event. Mean ALSFRS-R decline did not differ between groups (p = .69). Change in measured home time was similar between groups. Digital and in-clinic ALSFRS-R correlated well. DISCUSSION Inosine met pre-specified criteria for safety and tolerability. A functional benefit was not demonstrated in this trial designed for safety and tolerability. Findings suggested potential utility for a smartphone application in ALS clinical and research settings.
Collapse
Affiliation(s)
- David Walk
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Katharine Nicholson
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eduardo Locatelli
- Department of Neurology, Holy Cross Hospital, Fort Lauderdale, Florida, USA
| | - James Chan
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eric A Macklin
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Valerie Ferment
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Georgios Manousakis
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Marianne Chase
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mariah Connolly
- Clinical Research Organization, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Derek Dagostino
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Meghan Hall
- Clinical Research Organization, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Joseph Ostrow
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lindsay Pothier
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Cassandra Lieberman
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dario Gelevski
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rebecca Randall
- Clinical Research Organization, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Alexander V Sherman
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erin Steinhart
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniela Grasso Walker
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jason Walker
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hong Yu
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anne-Marie Wills
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael A Schwarzschild
- Department of Neurology, Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Boston, Massachusetts, USA
| | - Anna L Beukenhorst
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jukka-Pekka Onnela
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - James D Berry
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Merit E Cudkowicz
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sabrina Paganoni
- Neurological Clinical Research Institute and Healey & AMG Center for ALS, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Zhao Y, Liu X, Yang G. Adenosinergic Pathway in Parkinson's Disease: Recent Advances and Therapeutic Perspective. Mol Neurobiol 2023; 60:3054-3070. [PMID: 36786912 DOI: 10.1007/s12035-023-03257-3] [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: 06/04/2022] [Accepted: 02/07/2023] [Indexed: 02/15/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized pathologically by α-synuclein (α-syn) aggregation. In PD, the current mainstay of symptomatic treatment is levodopa (L-DOPA)-based dopamine (DA) replacement therapy. However, the development of dyskinesia and/or motor fluctuations which is relevant to levodopa is restricting its long-term utility. Given that the ability of which is to modulate the striato-thalamo-cortical loops and function to modulate basal ganglia output, the adenosinergic pathway (AP) is qualified as a potential promising non-DA target. As an indispensable component of energy production pathways, AP modulates cellular metabolism and gene regulation in both neurons and neuroglia cells through the recognition and degradation of extracellular adenosine. In addition, AP is geared to the initiation, evolution, and resolution of inflammation as well. Besides the above-mentioned crosstalk between the adenosine and dopamine signaling pathways, the functions of adenosine receptors (A1R, A2AR, A2BR, and A3R) and metabolism enzymes in modulating PD pathological process have been extensively investigated in recent decades. Here we reviewed the emerging findings focused on the function of adenosine receptors, adenosine formation, and metabolism in the brain and discussed its potential roles in PD pathological process. We also recapitulated clinical studies and the preclinical evidence for the medical strategies targeting the Ado signaling pathway to improve motor dysfunction and alleviate pathogenic process in PD. We hope that further clinical studies should consider this pathway in their monotherapy and combination therapy, which would open new vistas to more targeted therapeutic approaches.
Collapse
Affiliation(s)
- Yuan Zhao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Xin Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Guofeng Yang
- Department of Geriatrics, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, People's Republic of China. .,Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
22
|
Lenka A, Jankovic J. How should future clinical trials be designed in the search for disease-modifying therapies for Parkinson's disease? Expert Rev Neurother 2023; 23:107-122. [PMID: 36803618 DOI: 10.1080/14737175.2023.2177535] [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: 02/22/2023]
Abstract
INTRODUCTION Although there has been substantial progress in research and innovations in symptomatic treatments, similar success has not been achieved in disease-modifying therapy (DMT) for Parkinson's disease (PD). Considering the enormous motor, psychosocial and financial burden associated with PD, safe and effective DMT is of paramount importance. AREAS COVERED One of the reasons for the lack of progress in DMT for PD is poor or inappropriate design of clinical trials. In the first part of the article, the authors focus on the plausible reasons why the previous trials have failed and in the latter part, they provide their perspectives on future DMT trials. EXPERT OPINION There are several potential reasons why previous trials have failed, including broad clinical and etiopathogenic heterogeneity of PD, poor definition and documentation of target engagement, lack of appropriate biomarkers and outcome measures, and short duration of follow-up. To address these deficiencies, future trials may consider- (i) a more customized approach to select the most suitable participants and therapeutic approaches, (ii) explore combination therapies that would target multiple pathogenetic mechanisms, and (iii) moving beyond targeting only motor symptoms to also assessing non-motor features of PD in well-designed longitudinal studies.
Collapse
Affiliation(s)
- Abhishek Lenka
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
23
|
Muzio L, Ghirelli A, Agosta F, Martino G. Novel therapeutic approaches for motor neuron disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:523-537. [PMID: 37620088 DOI: 10.1016/b978-0-323-98817-9.00027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that leads to the neurodegeneration and death of upper and lower motor neurons (MNs). Although MNs are the main cells involved in the process of neurodegeneration, a growing body of evidence points toward other cell types as concurrent to disease initiation and propagation. Given the current absence of effective therapies, the quest for other therapeutic targets remains open and still challenges the scientific community. Both neuronal and extra-neuronal mechanisms of cellular stress and damage have been studied and have posed the basis for the development of novel therapies that have been investigated on both animal models and humans. In this chapter, a thorough review of the main mechanisms of cellular damage and the respective therapeutic attempts targeting them is reported. The main areas covered include neuroinflammation, protein aggregation, RNA metabolism, and oxidative stress.
Collapse
Affiliation(s)
- Luca Muzio
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy
| | - Alma Ghirelli
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Agosta
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Gianvito Martino
- San Raffaele Scientific Institute, Division of Neuroscience, InsPE, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
24
|
Song Y, Racette BA, Camacho-Soto A, Searles Nielsen S. Biologic targets of prescription medications and risk of neurodegenerative disease in United States Medicare beneficiaries. PLoS One 2023; 18:e0285011. [PMID: 37195983 DOI: 10.1371/journal.pone.0285011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
OBJECTIVE To identify prescription medications associated with a lower risk of three neurodegenerative diseases: Parkinson disease, Alzheimer disease, and amyotrophic lateral sclerosis. METHODS We conducted a population-based, case-control study of U.S. Medicare beneficiaries in 2009 (42,885 incident neurodegenerative disease cases, 334,387 randomly selected controls). Using medication data from 2006-2007, we categorized all filled medications according to their biological targets and mechanisms of action on those targets. We used multinomial logistic regression models, while accounting for demographics, indicators of smoking, and health care utilization, to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for 141 target-action pairs and each neurodegenerative disease. For target-action pairs inversely associated with all three diseases, we attempted replication in a cohort study that included an active comparator group. We constructed the cohort by following controls forward for incident neurodegenerative disease from the beginning of 2010 until death or end of 2014, i.e., up to five years after the two-year exposure lag. We used Cox proportional hazards regression while accounting for the same covariates. RESULTS The most consistent inverse association across both studies and all three neurodegenerative diseases was for xanthine dehydrogenase/oxidase blockers, represented by the gout medication, allopurinol. Allopurinol was associated with a 13-34% lower risk for each neurodegenerative disease group in multinomial regression, and a mean reduction of 23% overall, as compared to individuals who did not use allopurinol. In the replication cohort we observed a significant 23% reduction for neurodegenerative disease in the fifth year of follow-up, when comparing allopurinol users to non-users, and more marked associations with an active comparator group. We observed parallel associations for a related target-action pair unique to carvedilol. DISCUSSION/CONCLUSION Xanthine dehydrogenase/oxidase blockade might reduce risk of neurodegenerative disease. However, further research will be necessary to confirm that the associations related to this pathway are causal or to examine whether this mechanism slows progression.
Collapse
Affiliation(s)
- Yizhe Song
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Brad A Racette
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, United States of America
- Faculty of Health Sciences, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Alejandra Camacho-Soto
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Orthopedics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Susan Searles Nielsen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
25
|
Zeissler ML, McFarthing K, Raphael KG, Rafaloff G, Windle R, Carroll CB. An International Multi-Stakeholder Delphi Survey Study on the Design of Disease Modifying Parkinson's Disease Trials. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1343-1356. [PMID: 38007672 PMCID: PMC10741330 DOI: 10.3233/jpd-230109] [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: 10/23/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Design of disease modification (DM) trials for Parkinson's disease (PD) is challenging. Successful delivery requires a shared understanding of priorities and practicalities. OBJECTIVE To seek stakeholder consensus on phase 3 trials' overall goals and structure, inclusion criteria, outcome measures, and trial delivery and understand where perspectives differ. METHODS An international expert panel comprising people with Parkinson's (PwP), care partners (CP), clinical scientists, representatives from industry, funders and regulators participated in a survey-based Delphi study. Survey items were informed by a scoping review of DM trials and PwP input. Respondents scored item agreement over 3 rounds. Scores and reasoning were summarized by participant group each round until consensus, defined as≥70% of at least 3 participant groups falling within the same 3-point region of a 9-point Likert scale. RESULTS 92/121 individuals from 13 countries (46/69 PwP, 13/18 CP, 20/20 clinical scientists, representatives from 8/8 companies, 4/5 funders, and 1/1 regulator) completed the study. Consensus was reached on 14/31 survey items: 5/8 overall goals and structure, 1/8 Eligibility criteria, 7/13 outcome measures, and 1/2 trial delivery items. Extent of stakeholder endorsement for 428 reasons for scores was collated across items. CONCLUSIONS This is the first systematic multi-stakeholder consultation generating a unique repository of perspectives on pivotal aspects of DM trial design including those of PwP and CP. The panel endorsed outcomes that holistically measure PD and the importance of inclusive trials with hybrid delivery models. Areas of disagreement will inform mitigating strategies of researchers to ensure successful delivery of future trials.
Collapse
Affiliation(s)
| | | | - Karen G. Raphael
- College of Dentistry, New York University, New York, NY, USA
- Parkinson’s Research Advocate, USA
| | | | | | - Camille B. Carroll
- Faculty of Health, University of Plymouth, Plymouth, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| |
Collapse
|
26
|
Soares P, Silva C, Chavarria D, Silva FSG, Oliveira PJ, Borges F. Drug discovery and amyotrophic lateral sclerosis: Emerging challenges and therapeutic opportunities. Ageing Res Rev 2023; 83:101790. [PMID: 36402404 DOI: 10.1016/j.arr.2022.101790] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the degeneration of upper and lower motor neurons (MNs) leading to paralysis and, ultimately, death by respiratory failure 3-5 years after diagnosis. Edaravone and Riluzole, the only drugs currently approved for ALS treatment, only provide mild symptomatic relief to patients. Extraordinary progress in understanding the biology of ALS provided new grounds for drug discovery. Over the last two decades, mitochondria and oxidative stress (OS), iron metabolism and ferroptosis, and the major regulators of hypoxia and inflammation - HIF and NF-κB - emerged as promising targets for ALS therapeutic intervention. In this review, we focused our attention on these targets to outline and discuss current advances in ALS drug development. Based on the challenges and the roadblocks, we believe that the rational design of multi-target ligands able to modulate the complex network of events behind the disease can provide effective therapies in a foreseeable future.
Collapse
Affiliation(s)
- Pedro Soares
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| | - Catia Silva
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Filomena S G Silva
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paulo J Oliveira
- CNC - CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; IIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
27
|
Kim IS, Jo EK. Inosine: A bioactive metabolite with multimodal actions in human diseases. Front Pharmacol 2022; 13:1043970. [PMID: 36467085 PMCID: PMC9708727 DOI: 10.3389/fphar.2022.1043970] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/02/2022] [Indexed: 08/04/2023] Open
Abstract
The nucleoside inosine is an essential metabolite for purine biosynthesis and degradation; it also acts as a bioactive molecule that regulates RNA editing, metabolic enzyme activity, and signaling pathways. As a result, inosine is emerging as a highly versatile bioactive compound and second messenger of signal transduction in cells with diverse functional abilities in different pathological states. Gut microbiota remodeling is closely associated with human disease pathogenesis and responses to dietary and medical supplementation. Recent studies have revealed a critical link between inosine and gut microbiota impacting anti-tumor, anti-inflammatory, and antimicrobial responses in a context-dependent manner. In this review, we summarize the latest progress in our understanding of the mechanistic function of inosine, to unravel its immunomodulatory actions in pathological settings such as cancer, infection, inflammation, and cardiovascular and neurological diseases. We also highlight the role of gut microbiota in connection with inosine metabolism in different pathophysiological conditions. A more thorough understanding of the mechanistic roles of inosine and how it regulates disease pathologies will pave the way for future development of therapeutic and preventive modalities for various human diseases.
Collapse
Affiliation(s)
- In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, South Korea
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Eun-Kyoung Jo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, South Korea
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, South Korea
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon, South Korea
| |
Collapse
|
28
|
Seifar F, Dinasarapu AR, Jinnah HA. Uric Acid in Parkinson's Disease: What Is the Connection? Mov Disord 2022; 37:2173-2183. [PMID: 36056888 PMCID: PMC9669180 DOI: 10.1002/mds.29209] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/10/2022] Open
Abstract
Numerous studies have linked Parkinson's disease (PD) with low levels of uric acid (UA). Low UA has been associated with the risk of developing PD, and its progression and severity. The biological mechanisms underlying these relationships have never been firmly established. The most frequently proposed mechanism is that UA is an antioxidant. Low UA is thought to predispose to oxidative stress, which contributes to dopamine neuron degeneration, and leads to initial appearance of symptoms of PD and its worsening over time. Several recent studies have questioned this explanation. In this review, we describe the biology of UA, its many links with PD, evidence regarding UA as an antioxidant, and we question whether UA causes PD or contributes to its progression. We also address the possibility that something about PD causes low UA (reverse causation) or that low UA is a biomarker of some other more relevant mechanism in PD. We hope the evidence provided here will stimulate additional studies to better understand the links between UA and PD. Elucidating these mechanisms remains important, because they may provide new insights into the pathogenesis of PD or novel approaches to treatments. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Fatemeh Seifar
- Neurosciences Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta GA, USA
- Department of Neurology, Emory University, Atlanta GA, USA
| | | | - H. A. Jinnah
- Department of Neurology, Emory University, Atlanta GA, USA
- Department of Human Genetics, Emory University, Atlanta GA, USA
- Department of Pediatrics, Emory University, Atlanta GA, USA
| |
Collapse
|
29
|
Pou MA, Orfila F, Pagonabarraga J, Ferrer-Moret S, Corominas H, Diaz-Torne C. Risk of Parkinson's disease in a gout Mediterranean population: A case-control study. Joint Bone Spine 2022; 89:105402. [PMID: 35504516 DOI: 10.1016/j.jbspin.2022.105402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION High levels of serum urate has been associated to a neuroprotective effect in Parkinson's disease (PD) as an antioxidant agent. However, the relation between gout and PD remains contradictory. OBJECTIVE To study if the neuroprotective effect of serum urate is maintained in patients with gout in a large urban Mediterranean population. METHODS Primary care based matched case-control study, carried out using an electronic health record database from the public primary care health system of Barcelona. The database contains anonymous data from 1,520,934 patients. All patients, over 40 years old, with a new diagnostic record of PD, or a new prescription of dopaminergic drugs were included (incident cases). We randomly selected four controls for each case, matched by gender and age, with the frequency matching approach. Retrospective data of PD risk factors were also collected for each individual. A multivariate logistic regression model was used to evaluate the association of gout and PD, adjusted by the presence of other risk factors. RESULTS A new PD diagnosis was found in 17,629 individuals (incident diagnosis rate of 2.2 per 1000 individuals). Multivariate logistic regression model showed for gout: aOR=0.83 (0.76-0.91). When stratified by age, aOR for those under 75years was 0.99 (0.85-1.16) and 75 or over OR=0.77 (0.70-0.86). Dyslipidemia, hypertension and diabetes mellitus were associated with an increased risk of PD. Tobacco consumption was protective. CONCLUSION Our study, the first one made in a Mediterranean population, shows a PD protective effect of gout in both men and women over 75years old.
Collapse
Affiliation(s)
- Maria A Pou
- EAP Encants, Institut Català de la Salut, Barcelona, Spain
| | - Francesc Orfila
- Unitat de Suport a la Recerca, Ambit Barcelona Ciutat, Barcelona, Spain
| | | | | | - Hector Corominas
- Servei de Reumatologia, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cesar Diaz-Torne
- Servei de Reumatologia, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
30
|
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
|
31
|
Patterson CG, Joslin E, Gil AB, Spigle W, Nemet T, Chahine L, Christiansen CL, Melanson E, Kohrt WM, Mancini M, Josbeno D, Balfany K, Griffith G, Dunlap MK, Lamotte G, Suttman E, Larson D, Branson C, McKee KE, Goelz L, Poon C, Tilley B, Kang UJ, Tansey MG, Luthra N, Tanner CM, Haus JM, Fantuzzi G, McFarland NR, Gonzalez-Latapi P, Foroud T, Motl R, Schwarzschild MA, Simuni T, Marek K, Naito A, Lungu C, Corcos DM. Study in Parkinson's disease of exercise phase 3 (SPARX3): study protocol for a randomized controlled trial. Trials 2022; 23:855. [PMID: 36203214 PMCID: PMC9535216 DOI: 10.1186/s13063-022-06703-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND To date, no medication has slowed the progression of Parkinson's disease (PD). Preclinical, epidemiological, and experimental data on humans all support many benefits of endurance exercise among persons with PD. The key question is whether there is a definitive additional benefit of exercising at high intensity, in terms of slowing disease progression, beyond the well-documented benefit of endurance training on a treadmill for fitness, gait, and functional mobility. This study will determine the efficacy of high-intensity endurance exercise as first-line therapy for persons diagnosed with PD within 3 years, and untreated with symptomatic therapy at baseline. METHODS This is a multicenter, randomized, evaluator-blinded study of endurance exercise training. The exercise intervention will be delivered by treadmill at 2 doses over 18 months: moderate intensity (4 days/week for 30 min per session at 60-65% maximum heart rate) and high intensity (4 days/week for 30 min per session at 80-85% maximum heart rate). We will randomize 370 participants and follow them at multiple time points for 24 months. The primary outcome is the Movement Disorders Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) motor score (Part III) with the primary analysis assessing the change in MDS-UPDRS motor score (Part III) over 12 months, or until initiation of symptomatic antiparkinsonian treatment if before 12 months. Secondary outcomes are striatal dopamine transporter binding, 6-min walk distance, number of daily steps, cognitive function, physical fitness, quality of life, time to initiate dopaminergic medication, circulating levels of C-reactive protein (CRP), and brain-derived neurotrophic factor (BDNF). Tertiary outcomes are walking stride length and turning velocity. DISCUSSION SPARX3 is a Phase 3 clinical trial designed to determine the efficacy of high-intensity, endurance treadmill exercise to slow the progression of PD as measured by the MDS-UPDRS motor score. Establishing whether high-intensity endurance treadmill exercise can slow the progression of PD would mark a significant breakthrough in treating PD. It would have a meaningful impact on the quality of life of people with PD, their caregivers and public health. TRIAL REGISTRATION ClinicalTrials.gov NCT04284436 . Registered on February 25, 2020.
Collapse
Affiliation(s)
- Charity G. Patterson
- grid.21925.3d0000 0004 1936 9000Department of Physical Therapy, University of Pittsburgh, School of Health and Rehabilitation Sciences, 100 Technology Drive, Suite 500, Pittsburgh, PA 15219 USA
| | - Elizabeth Joslin
- grid.16753.360000 0001 2299 3507Department of Physical Therapy and Human Science, Northwestern University, Feinberg School of Medicine, Suite 1100, 645 North Michigan Avenue, Chicago, IL 60305 USA
| | - Alexandra B. Gil
- grid.21925.3d0000 0004 1936 9000Department of Physical Therapy, University of Pittsburgh, School of Health and Rehabilitation Sciences, 100 Technology Drive, Suite 500, Pittsburgh, PA 15219 USA
| | - Wendy Spigle
- grid.21925.3d0000 0004 1936 9000Department of Physical Therapy, University of Pittsburgh, School of Health and Rehabilitation Sciences, 100 Technology Drive, Suite 500, Pittsburgh, PA 15219 USA
| | - Todd Nemet
- grid.21925.3d0000 0004 1936 9000Department of Physical Therapy, University of Pittsburgh, School of Health and Rehabilitation Sciences, 100 Technology Drive, Suite 500, Pittsburgh, PA 15219 USA
| | - Lana Chahine
- grid.21925.3d0000 0004 1936 9000Department of Neurology, University of Pittsburgh, School of Medicine, 3471 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Cory L. Christiansen
- grid.430503.10000 0001 0703 675XDepartment of Physical Medicine & Rehabilitation, University of Colorado, School of Medicine, Aurora, CO 80217 USA
| | - Ed Melanson
- grid.430503.10000 0001 0703 675XDivision of Endocrinology, Metabolism and Diabetes, and Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.280930.0Eastern Colorado VA Health Care System, Geriatric Research Education and Clinical Center (GRECC), Denver, CO USA
| | - Wendy M. Kohrt
- grid.430503.10000 0001 0703 675XDivision of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.422100.50000 0000 9751 469XEastern Colorado Geriatric Research, Education, and Clinical Center, Rocky Mountain Regional VAMC, Aurora, USA
| | - Martina Mancini
- grid.5288.70000 0000 9758 5690Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Road, Portland, OR 97219 USA
| | - Deborah Josbeno
- grid.21925.3d0000 0004 1936 9000Department of Physical Therapy, University of Pittsburgh, School of Health and Rehabilitation Sciences, 100 Technology Drive, Suite 500, Pittsburgh, PA 15219 USA
| | - Katherine Balfany
- grid.430503.10000 0001 0703 675XDepartment of Physical Medicine & Rehabilitation, University of Colorado, School of Medicine, Aurora, CO 80217 USA
| | - Garett Griffith
- grid.16753.360000 0001 2299 3507Department of Physical Therapy and Human Science, Northwestern University, Feinberg School of Medicine, Suite 1100, 645 North Michigan Avenue, Chicago, IL 60305 USA
| | - Mac Kenzie Dunlap
- grid.239578.20000 0001 0675 4725Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195 USA
| | - Guillaume Lamotte
- grid.223827.e0000 0001 2193 0096Movement Disorders Division, Department of Neurology, University of Utah, 175 Medical Dr N, Salt Lake City, UT 84132 USA
| | - Erin Suttman
- grid.223827.e0000 0001 2193 0096Department of Physical Therapy & Athletic Training, University of Utah, 520 Wakara Way, Salt Lake City, UT 84115 USA
| | - Danielle Larson
- grid.16753.360000 0001 2299 3507Department of Neurology, Feinberg School of Medicine, Northwestern University, Suite 115, 710 N Lake Shore Drive, Chicago, IL 60611 USA
| | - Chantale Branson
- grid.9001.80000 0001 2228 775XMorehouse School of Medicine, 720 Westview Dr SW, Atlanta, GA 30310 USA
| | - Kathleen E. McKee
- grid.420884.20000 0004 0460 774XNeurosciences Clinical Program, Intermountain Healthcare, 5171 S Cottonwood Street, Suite 810, Murray, UT 84107 USA
| | - Li Goelz
- grid.185648.60000 0001 2175 0319Department of Kinesiology and Nutrition, UIC College of Applied Health Sciences, 919 W Taylor Street, Chicago, IL 60612 USA
| | - Cynthia Poon
- grid.16753.360000 0001 2299 3507Department of Neurology, Feinberg School of Medicine, Northwestern University, Suite 115, 710 N Lake Shore Drive, Chicago, IL 60611 USA
| | - Barbara Tilley
- grid.267308.80000 0000 9206 2401Department of Biostatistics and Data Science, University of Texas Health Science Center School of Public Health, 1200 Pressler Street E835, Houston, TX 77030 USA
| | - Un Jung Kang
- grid.240324.30000 0001 2109 4251NYU Langone Health, NYU Grossman School of Medicine, 435 E 30th Street, Science Building 1305, New York, NY 10016 USA
| | - Malú Gámez Tansey
- grid.15276.370000 0004 1936 8091Department of Neuroscience and Neurology, Normal Fixel Institute for Neurological Diseases and College of Medicine, University of Florida, 4911 Newell Road, Gainesville, FL 32610 USA
| | - Nijee Luthra
- grid.266102.10000 0001 2297 6811Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, 1651 4th Street, San Francisco, CA 94158 USA
| | - Caroline M. Tanner
- grid.266102.10000 0001 2297 6811Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, 1651 4th Street, San Francisco, CA 94158 USA
| | - Jacob M. Haus
- grid.214458.e0000000086837370School of Kinesiology, University of Michigan, 830 N. University Ave, Ann Arbor, MI 48109 USA
| | - Giamila Fantuzzi
- grid.185648.60000 0001 2175 0319Department of Kinesiology and Nutrition, UIC College of Applied Health Sciences, 919 W Taylor Street, Chicago, IL 60612 USA
| | - Nikolaus R. McFarland
- grid.15276.370000 0004 1936 8091Department of Neurology, Norman Fixel Institute for Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL 32608 USA
| | - Paulina Gonzalez-Latapi
- grid.16753.360000 0001 2299 3507Department of Neurology, Feinberg School of Medicine, Northwestern University, Suite 115, 710 N Lake Shore Drive, Chicago, IL 60611 USA
| | - Tatiana Foroud
- grid.257413.60000 0001 2287 3919Department of Medical and Molecular Genetics, Indiana University School of Medicine, 410 W. 10th Street, Indianapolis, IN 46220 USA
| | - Robert Motl
- grid.185648.60000 0001 2175 0319Department of Kinesiology and Nutrition, UIC College of Applied Health Sciences, 919 W Taylor Street, Chicago, IL 60612 USA
| | - Michael A. Schwarzschild
- grid.32224.350000 0004 0386 9924Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Rm 3002, 114 16th Street, Boston, MA 02129 USA
| | - Tanya Simuni
- grid.16753.360000 0001 2299 3507Department of Neurology, Feinberg School of Medicine, Northwestern University, Suite 115, 710 N Lake Shore Drive, Chicago, IL 60611 USA
| | - Kenneth Marek
- grid.429091.7Institute for Neurodegenerative Disorders, 60 Temple St, New Haven, CT 06510 USA
| | - Anna Naito
- grid.453428.c0000 0001 2236 2879Parkinson’s Foundation 200 SE 1st Street Suite 800, Miami, FL 33131 USA
| | - Codrin Lungu
- grid.416870.c0000 0001 2177 357XNational Institute of Neurological Disorders and Stroke, NIH, 6001 Executive Blvd, #2188, Rockville, MD 20852 USA
| | - Daniel M. Corcos
- grid.16753.360000 0001 2299 3507Department of Physical Therapy and Human Science, Northwestern University, Feinberg School of Medicine, Suite 1100, 645 North Michigan Avenue, Chicago, IL 60305 USA
| | | |
Collapse
|
32
|
Effects of elevated serum urate on cardiometabolic and kidney function markers in a randomised clinical trial of inosine supplementation. Sci Rep 2022; 12:12887. [PMID: 35902652 PMCID: PMC9334273 DOI: 10.1038/s41598-022-17257-1] [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: 03/10/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
In observational studies, serum urate positively associates with cardiometabolic and kidney diseases. We analyzed data from a randomised placebo-controlled trial to determine whether moderate hyperuricemia induced by inosine affects cardiometabolic and kidney function markers. One hundred and twenty post-menopausal women were recruited into a 6-month randomised, double-blind, placebo-controlled trial of inosine for bone health. Change from baseline in the following pre-specified endpoints was analyzed: body mass index; blood pressure; lipid profile; C-reactive protein; fasting glucose; insulin; HbA1c; serum creatinine; and estimated glomerular filtration rate (eGFR). Despite increases in serum urate levels (+ 0.17 mmol/L at week 6, P < 0.0001), no significant between-group differences were observed in cardiometabolic markers, with the exception of lower fasting glucose concentrations with inosine at week 19. In the inosine group, change in serum urate correlated with change in serum creatinine (r = 0.41, P = 0.0012). However, there was no between-group difference in serum creatinine values. Over the entire study period, there was no significant difference in eGFR (ANCOVA P = 0.13). Reduction in eGFR was greater in the inosine group at Week 13 (mean difference − 4.6 mL/min/1.73 m2, false detection rate P = 0.025), with no between-group difference in eGFR at other time points. These data indicate that increased serum urate does not negatively influence body mass index, blood pressure, lipid profile, or glycaemic control. Serum urate changes associated with inosine intake correlate with changes in serum creatinine, but this does not lead to clinically important reduction in kidney function over 6 months. Clinical trial registration number: Australia and New Zealand Clinical Trials Registry (ACTRN12617000940370), registered 30/06/2017.
Collapse
|
33
|
Basile MS, Bramanti P, Mazzon E. Inosine in Neurodegenerative Diseases: From the Bench to the Bedside. Molecules 2022; 27:molecules27144644. [PMID: 35889517 PMCID: PMC9316764 DOI: 10.3390/molecules27144644] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative diseases, such as Alzheimer′s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS), currently represent major unmet medical needs. Therefore, novel therapeutic strategies are needed in order to improve patients’ quality of life and prognosis. Since oxidative stress can be strongly involved in neurodegenerative diseases, the potential use of inosine, known for its antioxidant properties, in this context deserves particular attention. The protective action of inosine treatment could be mediated by its metabolite urate. Here, we review the current preclinical and clinical studies investigating the use of inosine in AD, PD, ALS, and MS. The most important properties of inosine seem to be its antioxidant action and its ability to raise urate levels and to increase energetic resources by improving ATP availability. Inosine appears to be generally safe and well tolerated; however, the possible formation of kidney stones should be monitored, and data on its effectiveness should be further explored since, so far, they have been controversial. Overall, inosine could be a promising potential strategy in the management of neurodegenerative diseases, and additional studies are needed in order to further investigate its safety and efficacy and its use as a complementary therapy along with other approved drugs.
Collapse
|
34
|
Wang S, Unnithan S, Bryant N, Chang A, Rosenthal LS, Pantelyat A, Dawson TM, Al‐Khalidi HR, West AB. Elevated Urinary Rab10 Phosphorylation in Idiopathic Parkinson Disease. Mov Disord 2022; 37:1454-1464. [PMID: 35521944 PMCID: PMC9308673 DOI: 10.1002/mds.29043] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pathogenic leucine-rich repeat kinase 2 LRRK2 mutations may increase LRRK2 kinase activity and Rab substrate phosphorylation. Genetic association studies link variation in LRRK2 to idiopathic Parkinson disease (iPD) risk. OBJECTIVES Through measurements of the LRRK2 kinase substrate pT73-Rab10 in urinary extracellular vesicles, this study seeks to understand how LRRK2 kinase activity might change with iPD progression. METHODS Using an immunoblotting approach validated in LRRK2 transgenic mice, the ratio of pT73-Rab10 to total Rab10 protein was measured in extracellular vesicles from a cross-section of G2019S LRRK2 mutation carriers (N = 45 participants) as well as 485 urine samples from a novel longitudinal cohort of iPD and controls (N = 85 participants). Generalized estimating equations were used to conduct analyses with commonly used clinical scales. RESULTS Although the G2019S LRRK2 mutation did not increase pT73-Rab10 levels, the ratio of pT73-Rab10 to total Rab10 nominally increased over baseline in iPD urine vesicle samples with time, but did not increase in age-matched controls (1.34-fold vs. 1.05-fold, 95% confidence interval [CI], 0.004-0.56; P = 0.046; Welch's t test). Effect estimates adjusting for sex, age, disease duration, diagnosis, and baseline clinical scores identified increasing total Movement Disorder Society-Sponsored Revision of the Unified (MDS-UPDRS) scores (β = 0.77; CI, 0.52-1.01; P = 0.0001) with each fold increase of pT73-Rab10 to total Rab10. Lower Montreal Cognitive Assessment (MoCA) score in iPD is also associated with increased pT73-Rab10. CONCLUSIONS These results provide initial insights into peripheral LRRK2-dependent Rab phosphorylation, measured in biobanked urine, where higher levels of pT73-Rab10 are associated with worse disease progression. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Shijie Wang
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| | - Shakthi Unnithan
- Department of Biostatistics and BioinformaticsDuke UniversityDurhamNorth CarolinaUSA
| | - Nicole Bryant
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| | - Allison Chang
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| | | | | | - Ted M. Dawson
- Department of NeurologyThe Johns Hopkins UniversityBaltimoreMarylandUSA
- Neurodegeneration and Stem Cell Programs, Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Solomon H. Snyder Department of NeuroscienceJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Hussein R. Al‐Khalidi
- Department of Biostatistics and BioinformaticsDuke UniversityDurhamNorth CarolinaUSA
| | - Andrew B. West
- Duke Center for Neurodegeneration and NeurotherapeuticsDuke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
35
|
Song Y, March J. Hyperuricemia and the small intestine: Transport mechanisms and co-morbidities. BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2022; 3:32-37. [PMID: 39416456 PMCID: PMC11446379 DOI: 10.1016/j.biotno.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 10/19/2024]
Abstract
There is a global increase in cases of hyperuricemia over the last 10 years. A critical component of serum uric acid control is the transport of uric acid to the intestinal lumen, which accounts for 30% of the uric acid eliminated from the serum. This mini review looks at two important aspects of elevated uric acid: the dynamics of intestinal uric acid transport and hyperuricemia co-morbidities. Elevated serum uric acid can lead to gout and it can also impact other diseases such as diabetes, cardiovascular diseases and nervous system diseases. The level of uric acid in the intestine could be related to the potential for uric acid to impact other morbidities. We review the evidence for this and what it would mean for persons with elevated serum uric acid.
Collapse
Affiliation(s)
- Yanbo Song
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
- Johnson and Johnson, China
| | - John March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
36
|
Combined drug triads for synergic neuroprotection in retinal degeneration. Biomed Pharmacother 2022; 149:112911. [DOI: 10.1016/j.biopha.2022.112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
|
37
|
Manzine PR, Vatanabe IP, Grigoli MM, Pedroso RV, de Almeida MPOMEP, de Oliveira DDSMS, Crispim Nascimento CM, Peron R, de Souza Orlandi F, Cominetti MR. Potential Protein Blood-Based Biomarkers in Different Types of Dementia: A Therapeutic Overview. Curr Pharm Des 2022; 28:1170-1186. [DOI: 10.2174/1381612828666220408124809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Biomarkers capable of identifying and distinguishing types of dementia such as Alzheimer's disease (AD), Parkinson's disease dementia (PDD), Lewy body dementia (LBD), and frontotemporal dementia (FTD) have been become increasingly relentless. Studies of possible biomarker proteins in the blood that can help formulate new diagnostic proposals and therapeutic visions of different types of dementia are needed. However, due to several limitations of these biomarkers, especially in discerning dementia, their clinical applications are still undetermined. Thus, the updating of biomarker blood proteins that can help in the diagnosis and discrimination of these main dementia conditions is essential to enable new pharmacological and clinical management strategies, with specificities for each type of dementia. To review the literature concerning protein blood-based AD and non-AD biomarkers as new pharmacological targets and/or therapeutic strategies. Recent findings for protein-based AD, PDD, LBD, and FTD biomarkers are focused on in this review. Protein biomarkers were classified according to the pathophysiology of the dementia types. The diagnosis and distinction of dementia through protein biomarkers is still a challenge. The lack of exclusive biomarkers for each type of dementia highlights the need for further studies in this field. Only after this, blood biomarkers may have a valid use in clinical practice as they are promising to help in diagnosis and in the differentiation of diseases.
Collapse
Affiliation(s)
- Patricia Regina Manzine
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Izabela Pereira Vatanabe
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Marina Mantellatto Grigoli
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Renata Valle Pedroso
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | | | | | | | - Rafaela Peron
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Fabiana de Souza Orlandi
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Márcia Regina Cominetti
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| |
Collapse
|
38
|
Dutta N, Deb I, Sarzynska J, Lahiri A. Inosine and its methyl derivatives: Occurrence, biogenesis, and function in RNA. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 169-170:21-52. [PMID: 35065168 DOI: 10.1016/j.pbiomolbio.2022.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/11/2021] [Accepted: 01/11/2022] [Indexed: 05/21/2023]
Abstract
Inosine is one of the most common post-transcriptional modifications. Since its discovery, it has been noted for its ability to contribute to non-Watson-Crick interactions within RNA. Rapidly accumulating evidence points to the widespread generation of inosine through hydrolytic deamination of adenosine to inosine by different classes of adenosine deaminases. Three naturally occurring methyl derivatives of inosine, i.e., 1-methylinosine, 2'-O-methylinosine and 1,2'-O-dimethylinosine are currently reported in RNA modification databases. These modifications are expected to lead to changes in the structure, folding, dynamics, stability and functions of RNA. The importance of the modifications is indicated by the strong conservation of the modifying enzymes across organisms. The structure, binding and catalytic mechanism of the adenosine deaminases have been well-studied, but the underlying mechanism of the catalytic reaction is not very clear yet. Here we extensively review the existing data on the occurrence, biogenesis and functions of inosine and its methyl derivatives in RNA. We also included the structural and thermodynamic aspects of these modifications in our review to provide a detailed and integrated discussion on the consequences of A-to-I editing in RNA and the contribution of different structural and thermodynamic studies in understanding its role in RNA. We also highlight the importance of further studies for a better understanding of the mechanisms of the different classes of deamination reactions. Further investigation of the structural and thermodynamic consequences and functions of these modifications in RNA should provide more useful information about their role in different diseases.
Collapse
Affiliation(s)
- Nivedita Dutta
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India
| | - Indrajit Deb
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India
| | - Joanna Sarzynska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland
| | - Ansuman Lahiri
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India.
| |
Collapse
|
39
|
Linetsky E, Abd Elhadi S, Bauer M, Gallant A, Namnah M, Weiss S, Segal D, Sharon R, Arkadir D. Safety and Tolerability, Dose-Escalating, Double-Blind Trial of Oral Mannitol in Parkinson's Disease. Front Neurol 2022; 12:716126. [PMID: 35046880 PMCID: PMC8761891 DOI: 10.3389/fneur.2021.716126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
Mannitol, a natural alcoholic-sugar, was recently suggested as a potential disease-modifying agent in Parkinson's disease. In animal models of the disease, mannitol interferes with the formation of α-synuclein fibrils, inhibits the formation of α-synuclein oligomers and leads to phenotypic recovery of impaired motor functions. Parkinson's patients who consume mannitol report improvements of both motor and non-motor symptoms. Safety of long-term use of oral mannitol, tolerable dose and possible benefit, however, were never clinically studied. We studied the safety of oral mannitol in Parkinson's disease and assessed the maximal tolerable oral dose by conducting a phase IIa, randomized, double-blind, placebo-controlled, single-center, dose-escalating study (ClinicalTrials.gov Identifier: NCT03823638). The study lasted 36 weeks and included four dose escalations of oral mannitol or dextrose to a maximal dose of 18 g per day. The primary outcome was the safety of oral mannitol, as assessed by the number of adverse events and abnormal laboratory results. Clinical and biochemical efficacy measures were collected but were not statistically-powered. Fourteen patients receiving mannitol completed the trial (in addition to eight patients on placebo). Mannitol-related severe adverse events were not observed. Gastrointestinal symptoms limited dose escalation in 6/14 participants on mannitol. None of the clinical or biochemical efficacy secondary outcome measures significantly differed between groups. We concluded that long-term use of 18 g per day of oral mannitol is safe in Parkinson's disease patients but only two third of patients tolerate this maximal dose. These findings should be considered in the design of future efficacy trials.
Collapse
Affiliation(s)
- Eduard Linetsky
- Department of Neurology, Faculty of Medicine, Hadassah Medical Organization, Hebrew University, Jerusalem, Israel
| | - Suaad Abd Elhadi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Max Bauer
- Department of Neurology, Faculty of Medicine, Hadassah Medical Organization, Hebrew University, Jerusalem, Israel
| | - Akiva Gallant
- Department of Neurology, Faculty of Medicine, Hadassah Medical Organization, Hebrew University, Jerusalem, Israel
| | - Montaser Namnah
- Department of Neurology, Faculty of Medicine, Hadassah Medical Organization, Hebrew University, Jerusalem, Israel
| | | | - Daniel Segal
- Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Sharon
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Arkadir
- Department of Neurology, Faculty of Medicine, Hadassah Medical Organization, Hebrew University, Jerusalem, Israel
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW This review aims to summarize recent evidence regarding the complex relationship between uric acid (UA), gout, and brain diseases. RECENT FINDINGS Observational studies have suggested that patients with hyperuricemia or gout might have a decreased risk of neurodegenerative diseases. Conversely, they may be at increased risk of cerebrovascular disease. Mendelian randomization (MR) studies use a genetic score as an instrumental variable to address the causality of the association between a risk factor (here, UA or gout) and an outcome. So far, MR analyses do not support a causal relationship of UA or gout with Alzheimer's disease and dementia, and of UA with Parkinson's disease or stroke. Observation studies indicate a U-shaped association between UA and brain diseases, but MR studies do not support that this association is causal. Further studies should address the causal role of gout as well as the impact of urate-lowering therapy on these outcomes.
Collapse
|
41
|
[Neuroprotective treatment of idiopathic, genetic and atypical Parkinson's disease with alpha-synuclein-Pathology]. DER NERVENARZT 2021; 92:1249-1259. [PMID: 34735584 PMCID: PMC8648656 DOI: 10.1007/s00115-021-01220-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 11/23/2022]
Abstract
Kernpunkt der Klassifikation neurodegenerativer Erkrankungen ist der histopathologische Nachweis von Ablagerungen bestimmter Proteine im Gehirn. Hierbei unterscheiden sich die verschiedenen Krankheitsentitäten sowohl hinsichtlich der Art der nachweisbaren Proteine als auch hinsichtlich der Konfiguration und Lokalisation der entsprechenden Proteinaggregate. Gemeinsames Kernmerkmal der als Synukleinopathien zusammengefassten Erkrankungen sind Ablagerungen des Proteins α‑Synuklein (ASYN). Die bekanntesten Erkrankungen dieses Spektrums sind die Parkinson-Krankheit (PK) mit neuronalem Nachweis von Lewy-Körperchen, die Demenz vom Lewy-Körper-Typ (DLK) mit zusätzlichem Nachweis von β‑Amyloid-Ablagerungen sowie die seltene Multisystematrophie (MSA) mit glialem Nachweis sog. Papp-Lantos-Körperchen. Da neben der diagnostischen mittlerweile auch die zentrale pathophysiologische Bedeutung des ASYN erwiesen ist, fokussiert sich die Entwicklung neuer Therapien aktuell auf die Beeinflussung der toxischen Wirkung dieses Proteins. Die verschiedenen Therapiekonzepte lassen sich grob in sechs Gruppen zusammenfassen: 1. die Verringerung der ASYN-Expression (Antisense-Therapie), 2. die Verhinderung der Bildung toxischer ASYN-Aggregate (Antiaggregativa, Chelatoren), 3. das Auflösen bzw. die Beseitigung intra- oder extrazellulärer toxischer ASYN-Aggregate (aktive und passive Immuntherapie, Antiaggregativa), 4. die Verstärkung zellulärer Abräummechanismen (Autophagie, lysosomale Mikrophagie) zur Beseitigung toxischer Formen von α‑Synuklein, 5. die Modulation neuroinflammatorischer Prozesse sowie 6. neuroprotektive Strategien. In diesem Artikel fassen wir die aktuellen Therapieentwicklungen zusammen und geben einen Ausblick auf vielversprechende zukünftige Therapieansätze.
Collapse
|
42
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
43
|
Protective Effects of Inosine on Memory Consolidation in a Rat Model of Scopolamine-Induced Cognitive Impairment: Involvement of Cholinergic Signaling, Redox Status, and Ion Pump Activities. Neurochem Res 2021; 47:446-460. [PMID: 34623562 DOI: 10.1007/s11064-021-03460-5] [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: 06/08/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 02/06/2023]
Abstract
This study investigated the effects of inosine on memory acquisition and consolidation, cholinesterases activities, redox status and Na+, K+-ATPase activity in a rat model of scopolamine-induced cognitive impairment. Adult male rats were divided into four groups: control (saline), scopolamine (1 mg/kg), scopolamine plus inosine (50 mg/kg), and scopolamine plus inosine (100 mg/kg). Inosine was pre-administered for 7 days, intraperitoneally. On day 8, scopolamine was administered pre (memory acquisition protocol) or post training (memory consolidation protocol) on inhibitory avoidance tasks. The animals were subjected to the step-down inhibitory avoidance task 24 hours after the training. Scopolamine induced impairment in the acquisition and consolidation phases; however, inosine was able to prevent only the impairment in memory consolidation. Also, scopolamine increased the activity of acetylcholinesterase and reduced the activity of Na+, K+-ATPase and the treatment with inosine protected against these alterations in consolidation protocol. In the animals treated with scopolamine, inosine improved the redox status by reducing the levels of reactive oxygen species and thiobarbituric acid reactive substances and restoring the activity of the antioxidant enzymes, superoxide dismutase and catalase. Our findings suggest that inosine may offer protection against scopolamine-induced memory consolidation impairment by modulating brain redox status, cholinergic signaling and ion pump activity. This compound may provide an interesting approach in pharmacotherapy and as a prophylactic against neurodegenerative mechanisms involved in Alzheimer's disease.
Collapse
|
44
|
Liu X, Teng T, Li X, Fan L, Xiang Y, Jiang Y, Du K, Zhang Y, Zhou X, Xie P. Impact of Inosine on Chronic Unpredictable Mild Stress-Induced Depressive and Anxiety-Like Behaviors With the Alteration of Gut Microbiota. Front Cell Infect Microbiol 2021; 11:697640. [PMID: 34595128 PMCID: PMC8476956 DOI: 10.3389/fcimb.2021.697640] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Current antidepressants do not confer a clear advantage in children and adolescents with major depressive disorder (MDD). Accumulating evidence highlights the potential antidepressant-like effects of inosine on adult MDD, and gut microbiomes are significantly associated with MDD via the microbiota-gut-brain axis. However, few studies have investigated possible associations between inosine and gut microbiota in adolescents with MDD. The current study investigated the potential antidepressant effects of inosine in adolescent male C57BL/6 mice. After 4 weeks of chronic unpredictable mild stress (CUMS) stimulation, the mice were assessed by body weight, the sucrose preference test (SPT), open field test, and the elevated plus maze (EPM). The microbiota compositions of feces were determined by 16S rRNA gene sequencing. Inosine significantly improved CUMS-induced depressive and anxiety-like behaviors in adolescent mice including SPT and EPM results. Fecal microbial composition differed in the CON+saline, CUMS+saline, and CUMS+inosine groups, which were characterized by 126 discriminative amplicon sequence variants belonging to Bacteroidetes and Firmicute at the phylum level and Muribaculaceae and Lachnospiraceae at the family level. Muribaculaceae was positively associated with depressive and anxiety-like behaviors. KEGG functional analysis suggested that inosine might affect gut microbiota through carbohydrate metabolism and lipid metabolism pathways. The results of the study indicated that inosine improved depressive and anxiety-like behaviors in adolescent mice, in conjunction with the alteration of fecal microbial composition. Our findings may provide a novel perspective on the antidepressant effects of inosine in children and adolescents.
Collapse
Affiliation(s)
- Xueer Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Teng Teng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Xuemei Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Li Fan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Yajie Xiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Yuanliang Jiang
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kang Du
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuqing Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyu Zhou
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Neurobiology, Chongqing, China
| |
Collapse
|
45
|
Grażyńska A, Adamczewska K, Antoniuk S, Bień M, Toś M, Kufel J, Urbaś W, Siuda J. The Influence of Serum Uric Acid Level on Non-Motor Symptoms Occurrence and Severity in Patients with Idiopathic Parkinson's Disease and Atypical Parkinsonisms-A Systematic Review. MEDICINA-LITHUANIA 2021; 57:medicina57090972. [PMID: 34577895 PMCID: PMC8468188 DOI: 10.3390/medicina57090972] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 12/31/2022]
Abstract
Background and Objectives: A growing number of studies correlated higher levels of serum uric acid (UA) with both: lower risk of Parkinson’s Disease (PD) occurrence and slower progression of the disease. Similar conclusions were made where studies correlated UA with atypical Parkinsonisms (AP) progression. A few researchers have studied the issue of the influence of serum UA on the occurrence of non-motor symptoms (NMS) in PD and AP. Our systematic review is the first review completely dedicated to this matter. Materials and Methods: A comprehensive evaluation of the literature was performed to review the relationship between UA and NMS in PD and AP. The systematic review was conducted according to PRISMA Statement guidelines. The following databases were searched starting in April 2021: MEDLINE via PubMed, Embase, and Scopus. During the research, the following filters were used: >2010, articles in English, concerning humans. The study was not registered and received no external funding. Results: Seven articles meeting all inclusion criteria were included in this study. Collectively, data on 1104 patients were analyzed. A correlation between serum UA concentration and a few NMS types has been provided by the analyzed studies. In four papers, sleep disorders and fatigue were related to UA for both advanced and early PD. Other commonly appearing NMS domains were Attention/memory (4 studies), Depression/anxiety (3 studies), Cardiovascular (3 studies), Gastrointestinal (1 study), Perceptual (1 study), and Miscellaneous (1 study). For AP, no significant correlation between UA and worsening of NMS has been found. Conclusions: Based on the analyzed studies, a correlation between serum UA level and the occurrence and worsening of NMS in PD and APs cannot be definitively determined. Large-scale studies with a more diverse patient population and with more accurate methods of NMS assessment in Parkinsonism are needed.
Collapse
Affiliation(s)
- Anna Grażyńska
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (A.G.); (K.A.); (S.A.); (M.B.)
| | - Klaudia Adamczewska
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (A.G.); (K.A.); (S.A.); (M.B.)
| | - Sofija Antoniuk
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (A.G.); (K.A.); (S.A.); (M.B.)
| | - Martyna Bień
- Students’ Scientific Association, Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland; (A.G.); (K.A.); (S.A.); (M.B.)
| | - Mateusz Toś
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Jakub Kufel
- Department of Biophysics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Zabrze, 41-800 Zabrze, Poland;
| | - Weronika Urbaś
- Department of Neurology, St. Barbara Provincial Specialist Hospital No. 5, 41-200 Sosnowiec, Poland;
| | - Joanna Siuda
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
- Correspondence: ; Tel.: +48-32-789-46-01 or +48-501-252-691; Fax: +48-32-789-45-55
| |
Collapse
|
46
|
Schwarzschild MA, Ascherio A, Casaceli C, Curhan GC, Fitzgerald R, Kamp C, Lungu C, Macklin EA, Marek K, Mozaffarian D, Oakes D, Rudolph A, Shoulson I, Videnovic A, Scott B, Gauger L, Aldred J, Bixby M, Ciccarello J, Gunzler SA, Henchcliffe C, Brodsky M, Keith K, Hauser RA, Goetz C, LeDoux MS, Hinson V, Kumar R, Espay AJ, Jimenez-Shahed J, Hunter C, Christine C, Daley A, Leehey M, de Marcaida JA, Friedman JH, Hung A, Bwala G, Litvan I, Simon DK, Simuni T, Poon C, Schiess MC, Chou K, Park A, Bhatti D, Peterson C, Criswell SR, Rosenthal L, Durphy J, Shill HA, Mehta SH, Ahmed A, Deik AF, Fang JY, Stover N, Zhang L, Dewey RB, Gerald A, Boyd JT, Houston E, Suski V, Mosovsky S, Cloud L, Shah BB, Saint-Hilaire M, James R, Zauber SE, Reich S, Shprecher D, Pahwa R, Langhammer A, LaFaver K, LeWitt PA, Kaminski P, Goudreau J, Russell D, Houghton DJ, Laroche A, Thomas K, McGraw M, Mari Z, Serrano C, Blindauer K, Rabin M, Kurlan R, Morgan JC, Soileau M, Ainslie M, Bodis-Wollner I, Schneider RB, Waters C, Ratel AS, Beck CA, Bolger P, Callahan KF, Crotty GF, Klements D, Kostrzebski M, McMahon GM, Pothier L, Waikar SS, Lang A, Mestre T. Effect of Urate-Elevating Inosine on Early Parkinson Disease Progression: The SURE-PD3 Randomized Clinical Trial. JAMA 2021; 326:926-939. [PMID: 34519802 PMCID: PMC8441591 DOI: 10.1001/jama.2021.10207] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/05/2021] [Indexed: 01/13/2023]
Abstract
Importance Urate elevation, despite associations with crystallopathic, cardiovascular, and metabolic disorders, has been pursued as a potential disease-modifying strategy for Parkinson disease (PD) based on convergent biological, epidemiological, and clinical data. Objective To determine whether sustained urate-elevating treatment with the urate precursor inosine slows early PD progression. Design, Participants, and Setting Randomized, double-blind, placebo-controlled, phase 3 trial of oral inosine treatment in early PD. A total of 587 individuals consented, and 298 with PD not yet requiring dopaminergic medication, striatal dopamine transporter deficiency, and serum urate below the population median concentration (<5.8 mg/dL) were randomized between August 2016 and December 2017 at 58 US sites, and were followed up through June 2019. Interventions Inosine, dosed by blinded titration to increase serum urate concentrations to 7.1-8.0 mg/dL (n = 149) or matching placebo (n = 149) for up to 2 years. Main Outcomes and Measures The primary outcome was rate of change in the Movement Disorder Society Unified Parkinson Disease Rating Scale (MDS-UPDRS; parts I-III) total score (range, 0-236; higher scores indicate greater disability; minimum clinically important difference of 6.3 points) prior to dopaminergic drug therapy initiation. Secondary outcomes included serum urate to measure target engagement, adverse events to measure safety, and 29 efficacy measures of disability, quality of life, cognition, mood, autonomic function, and striatal dopamine transporter binding as a biomarker of neuronal integrity. Results Based on a prespecified interim futility analysis, the study closed early, with 273 (92%) of the randomized participants (49% women; mean age, 63 years) completing the study. Clinical progression rates were not significantly different between participants randomized to inosine (MDS-UPDRS score, 11.1 [95% CI, 9.7-12.6] points per year) and placebo (MDS-UPDRS score, 9.9 [95% CI, 8.4-11.3] points per year; difference, 1.26 [95% CI, -0.59 to 3.11] points per year; P = .18). Sustained elevation of serum urate by 2.03 mg/dL (from a baseline level of 4.6 mg/dL; 44% increase) occurred in the inosine group vs a 0.01-mg/dL change in serum urate in the placebo group (difference, 2.02 mg/dL [95% CI, 1.85-2.19 mg/dL]; P<.001). There were no significant differences for secondary efficacy outcomes including dopamine transporter binding loss. Participants randomized to inosine, compared with placebo, experienced fewer serious adverse events (7.4 vs 13.1 per 100 patient-years) but more kidney stones (7.0 vs 1.4 stones per 100 patient-years). Conclusions and Relevance Among patients recently diagnosed as having PD, treatment with inosine, compared with placebo, did not result in a significant difference in the rate of clinical disease progression. The findings do not support the use of inosine as a treatment for early PD. Trial Registration ClinicalTrials.gov Identifier: NCT02642393.
Collapse
Affiliation(s)
- Michael A Schwarzschild
- Mass General Institute for Neurodegenerative Disease, Boston, Massachusetts
- Massachusetts General Hospital, Boston
| | | | | | | | - Rebecca Fitzgerald
- Parkinson's Foundation Research Advocates, Parkinson's Foundation, New York, New York
| | | | - Codrin Lungu
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Eric A Macklin
- Massachusetts General Hospital, Boston
- Harvard Medical School, Boston, Massachusetts
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Dariush Mozaffarian
- Tufts School of Medicine and Division of Cardiology, Tufts Medical Center, Boston, Massachusetts
- Friedman School of Nutrition Science and Policy, Boston, Massachusetts
| | - David Oakes
- University of Rochester, Rochester, New York
| | | | - Ira Shoulson
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | | | | | | | - Jason Aldred
- Inland Northwest Research, Spokane, Washington
- Selkirk Neurology, Spokane, Washington
| | | | | | | | - Claire Henchcliffe
- University of California, Irvine
- Weill Cornell Medical College, New York, New York
| | | | | | | | | | | | | | - Rajeev Kumar
- Rocky Mountain Movement Disorders Center, Englewood, Colorado
| | | | | | | | | | | | | | | | | | | | | | | | - David K Simon
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Tanya Simuni
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Cynthia Poon
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mya C Schiess
- The University of Texas Health Science Center, Houston McGovern Medical School, Houston
| | | | - Ariane Park
- The Ohio State University Wexner Medical Center, Columbus
| | | | | | - Susan R Criswell
- Washington University School of Medicine in St Louis, St Louis, Missouri
| | | | | | - Holly A Shill
- Banner Sun Health Research Institute, Sun City, Arizona
- University of Arizona School of Medicine-Phoenix
| | | | | | | | - John Y Fang
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | - Ashley Gerald
- University of Texas Southwestern Medical Center, Dallas
| | | | | | | | | | - Leslie Cloud
- VCU Parkinson's & Movement Disorders Center, Richmond, Virginia
| | | | | | | | | | - Stephen Reich
- University of Maryland School of Medicine, Baltimore
| | - David Shprecher
- Banner Sun Health Research Institute, Sun City, Arizona
- University of Arizona School of Medicine-Phoenix
| | - Rajesh Pahwa
- University of Kansas Medical Center, Kansas City
| | | | - Kathrin LaFaver
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Peter A LeWitt
- Henry Ford Hospital-West Bloomfield, West Bloomfield Township, Michigan
| | - Patricia Kaminski
- Henry Ford Hospital-West Bloomfield, West Bloomfield Township, Michigan
| | | | | | | | | | - Karen Thomas
- Sentara Neurology Specialists, Norfolk, Virginia
| | - Martha McGraw
- Center for Movement Disorders and Neurodegenerative Disease, Northwestern Medicine/Central DuPage Hospital, Winfield, Illinois
| | - Zoltan Mari
- Cleveland Clinic-Las Vegas, Las Vegas, Nevada
| | | | | | - Marcie Rabin
- Atlantic Neuroscience Institute, Summit, New Jersey
| | - Roger Kurlan
- Atlantic Neuroscience Institute, Summit, New Jersey
| | | | - Michael Soileau
- Texas Movement Disorder Specialists, Georgetown
- Scott & White Healthcare/Texas A&M University, Temple
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Sushrut S Waikar
- Boston University School of Medicine, Boston, Massachusetts
- Boston Medical Center, Boston, Massachusetts
| | - Anthony Lang
- University of Toronto, Toronto, Ontario, Canada
- Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, Toronto, Ontario, Canada
| | | |
Collapse
|
47
|
Nascimento FP, Macedo-Júnior SJ, Lapa-Costa FR, Cezar-Dos-Santos F, Santos ARS. Inosine as a Tool to Understand and Treat Central Nervous System Disorders: A Neglected Actor? Front Neurosci 2021; 15:703783. [PMID: 34504414 PMCID: PMC8421806 DOI: 10.3389/fnins.2021.703783] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Since the 1970s, when ATP was identified as a co-transmitter in sympathetic and parasympathetic nerves, it and its active metabolite adenosine have been considered relevant signaling molecules in biological and pathological processes in the central nervous system (CNS). Meanwhile, inosine, a naturally occurring purine nucleoside formed by adenosine breakdown, was considered an inert adenosine metabolite and remained a neglected actor on the purinergic signaling scene in the CNS. However, this scenario began to change in the 1980s. In the last four decades, an extensive group of shreds of evidence has supported the importance of mediated effects by inosine in the CNS. Also, inosine was identified as a natural trigger of adenosine receptors. This evidence has shed light on the therapeutic potential of inosine on disease processes involved in neurological and psychiatric disorders. Here, we highlight the clinical and preclinical studies investigating the involvement of inosine in chronic pain, schizophrenia, epilepsy, depression, anxiety, and in neural regeneration and neurodegenerative diseases, such as Parkinson and Alzheimer. Thus, we hope that this review will strengthen the knowledge and stimulate more studies about the effects promoted by inosine in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Francisney Pinto Nascimento
- Programa de Pós-Graduação em Biociências, Laboratório de Neurofarmacologia Clínica, Faculdade de Medicina, Universidade Federal da Integração Latino-Americana, Foz do Iguaçu, Brazil
| | | | | | - Fernando Cezar-Dos-Santos
- Programa de Pós-Graduação em Biociências, Laboratório de Neurofarmacologia Clínica, Faculdade de Medicina, Universidade Federal da Integração Latino-Americana, Foz do Iguaçu, Brazil
| | - Adair R S Santos
- Programa de Pós-Graduação em Neurociências, Laboratório de Neurobiologia da Dor e Inflamação, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
48
|
Ciobanu AM, Ionita I, Buleandra M, David IG, Popa DE, Ciucu AA, Budisteanu M. Current advances in metabolomic studies on non-motor psychiatric manifestations of Parkinson's disease (Review). Exp Ther Med 2021; 22:1010. [PMID: 34345292 PMCID: PMC8311266 DOI: 10.3892/etm.2021.10443] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
Life expectancy has increased worldwide and, along with it, a greater prevalence of age-dependent disorders, chronic illnesses and comorbidities can be observed. In 2019, in both Europe and the Americas, dementias ranked 3rd among the top 10 causes of death. Parkinson's disease (PD) is the second most frequent type of neurodegenerative disease. In the last decades, globally, the number of people suffering from PD has more than doubled to over 6 million. Of all the neurological disorders, PD increased with the fastest rate. This troubling trend highlights the stringent need for accurate diagnostic biomarkers, especially in the early stages of the disease and to evaluate treatment response. To gain a broad and complex understanding of the recent advances in the '-omics' research fields, electronic databases such as PubMed, Google Academic, and Science Direct were searched for publications regarding metabolomic studies on PD to identify specific biomarkers for PD, and especially PD with associated psychiatric symptomatology. Discoveries in the fields of metagenomics, transcriptomics and proteomics, may lead to an improved comprehension of the metabolic pathways involved in disease etiology and progression and contribute to the discovery of novel therapeutic targets for effective treatment options.
Collapse
Affiliation(s)
- Adela Magdalena Ciobanu
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
- Department of Neurosciences, Discipline of Psychiatry, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ioana Ionita
- Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Psychiatric Hospital, 041914 Bucharest, Romania
| | - Mihaela Buleandra
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Iulia Gabriela David
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Dana Elena Popa
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Anton Alexandru Ciucu
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Magdalena Budisteanu
- Laboratory of Medical Genetics, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
- Department of Medical Genetics, Faculty of Medicine, ‘Titu Maiorescu’ University, 031593 Bucharest, Romania
- Psychiatry Research Laboratory, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| |
Collapse
|
49
|
Lungu C, Cedarbaum JM, Dawson TM, Dorsey ER, Faraco C, Federoff HJ, Fiske B, Fox R, Goldfine AM, Kieburtz K, Macklin EA, Matthews H, Rafaloff G, Saunders-Pullman R, Schor NF, Schwarzschild MA, Sieber BA, Simuni T, Surmeier DJ, Tamiz A, Werner MH, Wright CB, Wyse R. Seeking progress in disease modification in Parkinson disease. Parkinsonism Relat Disord 2021; 90:134-141. [PMID: 34561166 DOI: 10.1016/j.parkreldis.2021.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/18/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Disease modification in Parkinson disease (PD) has remained an elusive goal, in spite of large investments over several decades. Following a large meeting of experts, this review article discusses the state of the science, possible reasons for past PD trials' failures to demonstrate disease-modifying benefit, and potential solutions. METHODS The National Institute of Neurological Disorders and Stroke (NINDS) convened a meeting including leaders in the field and representatives of key stakeholder groups to discuss drug therapy with the goal of disease modification in PD. RESULTS Important lessons can be learned from previous attempts, as well as from other fields. The selection process for therapeutic targets and agents differs among various organizations committed to therapeutic development. The areas identified as critical to target in future research include the development of relevant biomarkers, refinements of the targeted patient populations, considerations of novel trial designs, and improving collaborations between all stakeholders. CONCLUSIONS We identify potential barriers to progress in disease modification for Parkinson's and propose a set of research priorities that may improve the likelihood of success.
Collapse
Affiliation(s)
- Codrin Lungu
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 6001 Executive Blvd, #2188, Rockville, MD, 20852, USA.
| | | | - Ted M Dawson
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - E Ray Dorsey
- University of Rochester Medical Center, Rochester, NY, USA
| | - Carlos Faraco
- Division of Clinical Research, NINDS, NIH, Bethesda, MD, USA
| | | | - Brian Fiske
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Robert Fox
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Karl Kieburtz
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | | | | | | | | | - Tanya Simuni
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dalton J Surmeier
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Amir Tamiz
- Division of Translational Research, NINDS, NIH, Bethesda, MD, USA
| | | | | | | |
Collapse
|
50
|
de Aquino CH. Methodological Issues in Randomized Clinical Trials for Prodromal Alzheimer's and Parkinson's Disease. Front Neurol 2021; 12:694329. [PMID: 34421799 PMCID: PMC8377160 DOI: 10.3389/fneur.2021.694329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/22/2021] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the first and second most common neurodegenerative disorders, respectively. Both are proteinopathies with inexorable courses and no approved disease-modifying therapies. A substantial effort has been made to identify interventions that could slow down the progression of AD and PD; to date, with no success. The advances in biomarker research improved the identification of individuals at risk for these disorders before symptom onset, recognizing the pre-clinical stage, in which there is abnormal protein accumulation but no clinical symptoms of the disease, and the prodromal stage, in which mild symptoms are present but the clinical diagnostic criteria for disease cannot be fulfilled. The ability to detect pre-clinical and prodromal stages of these diseases has encouraged clinical trials for disease-modification at earlier phases, seeking to slow or prevent phenoconversion into clinical disease. Clinical trials at these stages have several challenges, such as the identification of the eligible population, the appropriate choice of biomarkers, the definition of clinical endpoints, the duration of follow-up, and the statistical analysis. This article aims to discuss some of the methodological challenges in the design of trials for pre-clinical and prodromal phases of AD and PD, to critically review the recent studies, and to discuss methodological approaches to mitigate these challenges in trial design.
Collapse
Affiliation(s)
- Camila Henriques de Aquino
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.,Department of Health, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| |
Collapse
|