1
|
Li Q, Song Z, Peng L, Feng S, Zhan K, Ling H. Dihydromyricetin Improves High Glucose-Induced Dopaminergic Neuronal Damage by Activating AMPK-Autophagy Signaling Pathway. Exp Clin Endocrinol Diabetes 2024. [PMID: 39168148 DOI: 10.1055/a-2399-1174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
INTRODUCTION In recent years, a growing number of clinical and biological studies have shown that patients with type 2 diabetes mellitus (T2DM) are at increased risk of developing Parkinson's disease (PD). Prolonged exposure to hyperglycemia results in abnormal glucose metabolism, which in turn causes pathological changes similar to PD, leading to selective loss of dopaminergic neurons in the compact part of the substantia nigra. Dihydromyricetin (DHM) is a naturally occurring flavonoid with various biological activities including antioxidant and hepatoprotective properties. In this study, the effect of DHM on high glucose-induced dopaminergic neuronal damage was investigated. METHODS The potential modulatory effects of DHM on high glucose-induced dopaminergic neuronal damage and its mechanism were studied. RESULTS DHM ameliorated high glucose-induced dopaminergic neuronal damage and autophagy injury. Inhibition of autophagy by 3-methyladenine abrogated the beneficial effects of DHM on high glucose-induced dopaminergic neuronal damage. In addition, DHM increased levels of p-AMP-activated protein kinase (AMPK) and phosphorylated UNC51-like kinase 1. The AMPK inhibitor compound C eliminated DHM-induced autophagy and subsequently inhibited the ameliorative effects of DHM on high glucose-induced dopaminergic neuronal damage. DISCUSSION DHM ameliorates high glucose-induced dopaminergic neuronal damage by activating the AMPK-autophagy pathway.
Collapse
Affiliation(s)
- Qi Li
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
- Department of Pathology, Yuebei People's Hospital Affiliated to Shantou University School of Medicine, Shaoguan, China
| | - Zhenjiang Song
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
| | - Liting Peng
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
| | - Shuidong Feng
- Department of Social Medicine and Health Management, University of South China Hengyang Medical School, Hengyang, China
| | - Kebin Zhan
- Department of Neurology, The Second Affiliated Hospital, University of South China Hengyang Medical School, Hengyang, China
| | - Hongyan Ling
- Department of Physiology, University of South China Hengyang Medical School, Hengyang, China
| |
Collapse
|
2
|
Yoo HS, Kim HK, Lee HS, Yoon SH, Na HK, Kang SW, Lee JH, Ryu YH, Lyoo CH. Predictors associated with the rate of progression of nigrostriatal degeneration in Parkinson's disease. J Neurol 2024; 271:5213-5222. [PMID: 38839638 DOI: 10.1007/s00415-024-12477-z] [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: 12/20/2023] [Revised: 04/20/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Parkinson's disease (PD) manifests as a wide variety of clinical phenotypes and its progression varies greatly. However, the factors associated with different disease progression remain largely unknown. METHODS In this retrospective cohort study, we enrolled 113 patients who underwent 18F-FP-CIT PET scan twice. Given the negative exponential progression pattern of dopamine loss in PD, we applied the natural logarithm to the specific binding ratio (SBR) of two consecutive 18F-FP-CIT PET scans and conducted linear mixed model to calculate individual slope to define the progression rate of nigrostriatal degeneration. We investigated the clinical and dopamine transporter (DAT) availability patterns associated with the progression rate of dopamine depletion in each striatal sub-region. RESULTS More symmetric parkinsonism, the presence of dyslipidemia, lower K-MMSE total score, and lower anteroposterior gradient of the mean putaminal SBR were associated with faster progression rate of dopamine depletion in the caudate nucleus. More symmetric parkinsonism and lower anteroposterior gradient of the mean putaminal SBR were associated with faster depletion of dopamine in the anterior putamen. Older age at onset, more symmetric parkinsonism, the presence of dyslipidemia, and lower anteroposterior gradient of the mean putaminal SBR were associated with faster progression rate of dopamine depletion in the posterior putamen. Lower striatal mean SBR predicted the development of LID, while lower mean SBR in the caudate nuclei predicted the development of dementia. DISCUSSION Our results suggest that the evaluation of baseline clinical features and patterns of DAT availability can predict the progression of PD and its prognosis.
Collapse
Affiliation(s)
- Han Soo Yoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Han-Kyeol Kim
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - So Hoon Yoon
- Department of Neurology, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, South Korea
| | - Han Kyu Na
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Sung Woo Kang
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Jae-Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea.
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea.
| |
Collapse
|
3
|
Ren Q, Fu J, Duan X, Sun L, Mu Z, Liang W, Li Y, Wang Z, Xiu S. The Effects of Ketogenic Diet on Brain Gene Expressions in Type 2 Diabetes Background. Neuroscience 2024; 549:101-109. [PMID: 38734303 DOI: 10.1016/j.neuroscience.2024.04.008] [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: 02/07/2024] [Revised: 04/07/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a major risk factor of a number of neurodegenerative diseases (NDDs). Ketogenic diet (KD) has significant beneficial effects on glycemic control and may act effectively against NDDs, but the mechanism remains unclear. In this study, we aimed to investigate the potential effects of KD on gene expressions in the brains of T2DM model mice. Male db/db mice at the age of 9 weeks were fed with KD or normal diet to the age of 6 months, and the whole brains were subjected to mRNA-seq analysis for differentially expressed genes. KD significantly lowered fasting glucose and body weights in db/db mice (P < 0.05), and the expression of 189 genes in the brain were significantly changed (P < 0.05, |log2| > 1). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed that the differentially expressed genes upon KD are involved in inflammatory responses and the functions of biosynthesis. In inflammatory responses, NF-κB signaling pathway, viral protein interaction with cytokine and cytokine receptor, and cytokine-cytokine receptor interaction pathways were enriched, and in biosynthesis pathways, genes functioning in lipid and amino acid metabolism, protein synthesis, and energy metabolism were enriched. Moreover, consistent with the gene set enrichment analysis results, proteasomal activity measured biochemically were enhanced in KD-fed T2DM mice. These data may facilitate the understanding of how KD can be protective to the brain in T2DM background. KD could be a new strategy for the prevention of NDDs in T2DM patients.
Collapse
Affiliation(s)
- Qianxu Ren
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Junling Fu
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaoye Duan
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lina Sun
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zhijing Mu
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wenping Liang
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Department of Neurology, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Shuangling Xiu
- Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
4
|
Komici K, Pansini A, Bencivenga L, Rengo G, Pagano G, Guerra G. Frailty and Parkinson's disease: the role of diabetes mellitus. Front Med (Lausanne) 2024; 11:1377975. [PMID: 38882667 PMCID: PMC11177766 DOI: 10.3389/fmed.2024.1377975] [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: 01/28/2024] [Accepted: 05/02/2024] [Indexed: 06/18/2024] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease associated with a progressive loss of dopaminergic neurons, clinically characterized by motor and non-motor signs. Frailty is a clinical condition of increased vulnerability and negative health outcomes due to the loss of multiple physiological reserves. Chronic hyperglycemia and insulin resistance, which characterize diabetes mellitus (DM), have been reported to alter dopaminergic activity, increase the risk of PD, and influence the development of frailty. Even though diabetes may facilitate the development of frailty in patients with PD, this relationship is not established and a revision of the current knowledge is necessary. Furthermore, the synergy between DM, PD, and frailty may drive clinical complexity, worse outcomes, and under-representation of these populations in the research. In this review, we aimed to discuss the role of diabetes in the development of frailty among patients with PD. We summarized the clinical characteristics and outcomes of patients with concomitant DM, PD, and frailty. Finally, interventions to prevent frailty in this population are discussed.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
- Istituti Clinici Scientifici Maugeri IRCCS-Scientific Institute of Telese Terme, Telese Terme, BN, Italy
| | - Gennaro Pagano
- Roche Pharma Research and Early Development (pRED), Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
- University of Exeter Medical School, London, United Kingdom
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| |
Collapse
|
5
|
Lee KS, Yoon SH, Hwang I, Ma JH, Yang E, Kim RH, Kim E, Yu JW. Hyperglycemia enhances brain susceptibility to lipopolysaccharide-induced neuroinflammation via astrocyte reprogramming. J Neuroinflammation 2024; 21:137. [PMID: 38802820 PMCID: PMC11131277 DOI: 10.1186/s12974-024-03136-1] [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: 12/17/2023] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Hyperglycemia has been shown to modulate the immune response of peripheral immune cells and organs, but the impact of hyperglycemia on neuroinflammation within the brain remains elusive. In the present study, we provide evidences that streptozotocin (STZ)-induced hyperglycemic condition in mice drives a phenotypic switch of brain astrocytes to a proinflammatory state, and increases brain vulnerability to mild peripheral inflammation. In particular, we found that hyperglycemia led to a significant increase in the astrocyte proliferation as determined by flow cytometric and immunohistochemical analyses of mouse brain. The increased astrocyte proliferation by hyperglycemia was reduced by Glut1 inhibitor BAY-876. Transcriptomic analysis of isolated astrocytes from Aldh1l1CreERT2;tdTomato mice revealed that peripheral STZ injection induced astrocyte reprogramming into proliferative, and proinflammatory phenotype. Additionally, STZ-induced hyperglycemic condition significantly enhanced the infiltration of circulating myeloid cells into the brain and the disruption of blood-brain barrier in response to mild lipopolysaccharide (LPS) administration. Systemic hyperglycemia did not alter the intensity and sensitivity of peripheral inflammation in mice to LPS challenge, but increased the inflammatory potential of brain microglia. In line with findings from mouse experiments, a high-glucose environment intensified the LPS-triggered production of proinflammatory molecules in primary astrocyte cultures. Furthermore, hyperglycemic mice exhibited a significant impairment in cognitive function after mild LPS administration compared to normoglycemic mice as determined by novel object recognition and Y-maze tasks. Taken together, these results demonstrate that hyperglycemia directly induces astrocyte reprogramming towards a proliferative and proinflammatory phenotype, which potentiates mild LPS-triggered inflammation within brain parenchymal regions.
Collapse
Affiliation(s)
- Kyung-Seo Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Hyun Yoon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong-Hwa Ma
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Euimo Yang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Rebekah Hyeyoon Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eosu Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Kalinderi K, Papaliagkas V, Fidani L. GLP-1 Receptor Agonists: A New Treatment in Parkinson's Disease. Int J Mol Sci 2024; 25:3812. [PMID: 38612620 PMCID: PMC11011817 DOI: 10.3390/ijms25073812] [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: 02/13/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Recent data highlight similarities between neurodegenerative diseases, including PD and type 2 diabetes mellitus (T2DM), suggesting a crucial interplay between the gut-brain axis. Glucagon-like peptide-1 receptor (GLP-1R) agonists, known for their use in T2DM treatment, are currently extensively studied as novel PD modifying agents. For this narrative review article, we searched PubMed and Scopus databases for peer-reviewed research, review articles and clinical trials regarding GLP-1R agonists and PD published in the English language with no time restrictions. We also screened the references of the selected articles for possible additional articles in order to include most of the key recent evidence. Many data on animal models and preclinical studies show that GLP1-R agonists can restore dopamine levels, inhibit dopaminergic loss, attenuate neuronal degeneration and alleviate motor and non-motor features of PD. Evidence from clinical studies is also very promising, enhancing the possibility of adding GLP1-R agonists to the current armamentarium of drugs available for PD treatment.
Collapse
Affiliation(s)
- Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece;
| | - Liana Fidani
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
7
|
Visser AE, de Vries NM, Richard E, Bloem BR. Tackling vascular risk factors as a possible disease modifying intervention in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:50. [PMID: 38431725 PMCID: PMC10908840 DOI: 10.1038/s41531-024-00666-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Affiliation(s)
- Anne E Visser
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands.
- Department of Neurology, Haaglanden Medical Center, The Hague, The Netherlands.
| | - Nienke M de Vries
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Edo Richard
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Center of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Ahn JH, Kang MC, Youn J, Park KA, Han KD, Jung JH. Nonarteritic anterior ischemic optic neuropathy and incidence of Parkinson's disease based on a nationwide population based study. Sci Rep 2024; 14:2930. [PMID: 38316950 PMCID: PMC10844599 DOI: 10.1038/s41598-024-53196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024] Open
Abstract
This study aimed to investigate the association between nonarteritic anterior ischemic optic neuropathy (NAION) and Parkinson's disease (PD) using a retrospective, nationwide, population-based cohort in South Korea. This study utilized data from the Korean National Health Insurance database, including 43,960 NAION patients and 219,800 age- and sex-matched controls. Cox proportional hazards regression models were used to assess the risk of developing PD in the NAION group compared to the control group after adjusting for various confounding factors. Subgroup analyses were conducted based on sex, age, and comorbidities. The incidence rate of PD was higher in the NAION group (1.326 per 1000 person-years) than in the control group (0.859 per 1000 person-years). After adjusting for confounding factors, the risk of developing PD was significantly higher in the NAION group (adjusted hazard ratio [aHR] 1.516, 95% confidence interval [CI] 1.300-1.769). Subgroup analyses did not reveal a significant difference in the risk of PD development based on sex, age, or comorbidities. This retrospective, nationwide, population-based cohort study revealed a significant association between NAION and an increased risk of developing PD in a South Korean population. The incidence rate of PD was observed to be higher in individuals diagnosed with NAION than in age- and sex-matched controls even after adjusting for potential confounding variables, with the risk being approximately 51.6% higher in the NAION group. Further research is necessary to elucidate the underlying pathophysiological mechanisms linking NAION to PD and to determine whether similar associations exist in other ethnic and geographical populations.
Collapse
Affiliation(s)
- Jong Hyeon Ahn
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Min Chae Kang
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Jinyoung Youn
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
| | - Kyung-Ah Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Kyung-Do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Jin-Hyung Jung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
9
|
Greten S, Wegner F, Jensen I, Krey L, Rogozinski S, Fehring M, Heine J, Doll-Lee J, Pötter-Nerger M, Zeitzschel M, Hagena K, Pedrosa DJ, Eggers C, Bürk K, Trenkwalder C, Claus I, Warnecke T, Süß P, Winkler J, Gruber D, Gandor F, Berg D, Paschen S, Classen J, Pinkhardt EH, Kassubek J, Jost WH, Tönges L, Kühn AA, Schwarz J, Peters O, Dashti E, Priller J, Spruth EJ, Krause P, Spottke A, Schneider A, Beyle A, Kimmich O, Donix M, Haussmann R, Brandt M, Dinter E, Wiltfang J, Schott BH, Zerr I, Bähr M, Buerger K, Janowitz D, Perneczky R, Rauchmann BS, Weidinger E, Levin J, Katzdobler S, Düzel E, Glanz W, Teipel S, Kilimann I, Prudlo J, Gasser T, Brockmann K, Hoffmann DC, Klockgether T, Krause O, Heck J, Höglinger GU, Klietz M. The comorbidity and co-medication profile of patients with progressive supranuclear palsy. J Neurol 2024; 271:782-793. [PMID: 37803149 PMCID: PMC10827866 DOI: 10.1007/s00415-023-12006-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is usually diagnosed in elderly. Currently, little is known about comorbidities and the co-medication in these patients. OBJECTIVES To explore the pattern of comorbidities and co-medication in PSP patients according to the known different phenotypes and in comparison with patients without neurodegenerative disease. METHODS Cross-sectional data of PSP and patients without neurodegenerative diseases (non-ND) were collected from three German multicenter observational studies (DescribePSP, ProPSP and DANCER). The prevalence of comorbidities according to WHO ICD-10 classification and the prevalence of drugs administered according to WHO ATC system were analyzed. Potential drug-drug interactions were evaluated using AiDKlinik®. RESULTS In total, 335 PSP and 275 non-ND patients were included in this analysis. The prevalence of diseases of the circulatory and the nervous system was higher in PSP at first level of ICD-10. Dorsopathies, diabetes mellitus, other nutritional deficiencies and polyneuropathies were more frequent in PSP at second level of ICD-10. In particular, the summed prevalence of cardiovascular and cerebrovascular diseases was higher in PSP patients. More drugs were administered in the PSP group leading to a greater percentage of patients with polypharmacy. Accordingly, the prevalence of potential drug-drug interactions was higher in PSP patients, especially severe and moderate interactions. CONCLUSIONS PSP patients possess a characteristic profile of comorbidities, particularly diabetes and cardiovascular diseases. The eminent burden of comorbidities and resulting polypharmacy should be carefully considered when treating PSP patients.
Collapse
Affiliation(s)
- Stephan Greten
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Ida Jensen
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Lea Krey
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Sophia Rogozinski
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Meret Fehring
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Johanne Heine
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Johanna Doll-Lee
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Monika Pötter-Nerger
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Molly Zeitzschel
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Keno Hagena
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - David J Pedrosa
- Department of Neurology, University Hospital of Marburg and Gießen, 35043, BaldingerstraßeMarburg, Germany
| | - Carsten Eggers
- Department of Neurology, Knappschaftskrankenhaus Bottrop, Osterfelder Str. 157, 46242, Bottrop, Germany
| | - Katrin Bürk
- Kliniken Schmieder Stuttgart-Gerlingen, Solitudestraße 20, 70839, Gerlingen, Germany
| | | | - Inga Claus
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Tobias Warnecke
- Department of Neurology and Neurorehabilitation, Klinikum Osnabrueck-Academic Teaching Hospital of the WWU Muenster, Am Finkenhügel 1, 49076, Osnabrueck, Germany
| | - Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schloßplatz 4, 91054, Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schloßplatz 4, 91054, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schloßplatz 4, 91054, Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schloßplatz 4, 91054, Erlangen, Germany
| | - Doreen Gruber
- Movement Disorders Hospital, Beelitz-Heilstätten, Straße Nach Fichtenwalde 16, 14547, Beelitz-Heilstätten, Germany
| | - Florin Gandor
- Movement Disorders Hospital, Beelitz-Heilstätten, Straße Nach Fichtenwalde 16, 14547, Beelitz-Heilstätten, Germany
| | - Daniela Berg
- Department of Neurology, Kiel University, Christian-Albrechts-Platz 4, 24118, Kiel, Germany
| | - Steffen Paschen
- Department of Neurology, Kiel University, Christian-Albrechts-Platz 4, 24118, Kiel, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig Medical Center, Liebigstraße, 18, 04103, Leipzig, Germany
| | - Elmar H Pinkhardt
- Department of Neurology, University of Ulm, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Oberer Eselsberg 45, 89081, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Oberer Eselsberg, 89081, Ulm, Germany
| | - Wolfgang H Jost
- Parkinson-Klinik Ortenau, Kreuzbergstraße 12, 77709, Wolfach, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr University Bochum, Gudrunstraße 56, 44791, Bochum, Germany
- Protein Research Unit Ruhr (PURE), Neurodegeneration Research, Ruhr University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
| | - Andrea A Kühn
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité, University Medicine Berlin, Charitépl. 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Charitépl. 1, 10117, Berlin, Germany
| | - Johannes Schwarz
- Department of Neurology, Klinik Haag I. OB, Krankenhausstraße 1, 84453, Mühldorf a. Inn, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Charitépl. 1, 10117, Berlin, Germany
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Charitépl. 1, 10117, Berlin, Germany
| | - Eman Dashti
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitépl. 1, 10117, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Charitépl. 1, 10117, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitépl. 1, 10117, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Klinikum Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Eike J Spruth
- German Center for Neurodegenerative Diseases (DZNE), Charitépl. 1, 10117, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Charitépl. 1, 10117, Berlin, Germany
| | - Patricia Krause
- German Center for Neurodegenerative Diseases (DZNE), Charitépl. 1, 10117, Berlin, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Aline Beyle
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Okka Kimmich
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Markus Donix
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Robert Haussmann
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Moritz Brandt
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Elisabeth Dinter
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307, Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Mathias Bähr
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center, Georg August University, Von-Siebold-Str. 5, 37075, Göttingen, Germany
- Cluster of Excellence Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Daniel Janowitz
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, Exhibition Rd, South Kensington, London, SW7 2BX, UK
| | - Boris-Stephan Rauchmann
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Endy Weidinger
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Universitätspl. 2, 39106, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, Gower St, London, WC1E 6BT, UK
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, 39120, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Universitätspl. 2, 39106, Magdeburg, Germany
- Clinic for Neurology, Medical Faculty, University Hospital Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Straße 20, 18147, Rostock-GreifswaldRostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Schillingallee 35, 18057, Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Straße 20, 18147, Rostock-GreifswaldRostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Schillingallee 35, 18057, Rostock, Germany
| | - Johannes Prudlo
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Straße 20, 18147, Rostock-GreifswaldRostock, Germany
- Department of Neurology, University Medical Center, Schillingallee 35, 18057, Rostock, Germany
| | - Thomas Gasser
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, 72076, Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Kathrin Brockmann
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, 72076, Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Straße 3, 72076, Tübingen, Germany
| | - Daniel C Hoffmann
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Thomas Klockgether
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Neurology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Olaf Krause
- Center for Medicine of the Elderly, DIAKOVERE Henriettenstift and Department of General Medicine and Palliative Care, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- Center for Geriatric Medicine, Hospital DIAKOVERE Henriettenstift, Schwemannstrasse 19, 30559, Hannover, Germany
| | - Johannes Heck
- Institute for Clinical Pharmacology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Günter U Höglinger
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377, Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany
| | - Martin Klietz
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| |
Collapse
|
10
|
König A, Outeiro TF. Diabetes and Parkinson's Disease: Understanding Shared Molecular Mechanisms. JOURNAL OF PARKINSON'S DISEASE 2024; 14:917-924. [PMID: 38995799 PMCID: PMC11307096 DOI: 10.3233/jpd-230104] [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: 02/19/2024] [Indexed: 07/14/2024]
Abstract
Aging is a major risk factor for Parkinson's disease (PD). Genetic mutations account for a small percentage of cases and the majority appears to be sporadic, with yet unclear causes. However, various environmental factors have been linked to an increased risk of developing PD and, therefore, understanding the complex interplay between genetic and environmental factors is crucial for developing effective disease-modifying therapies. Several studies identified a connection between type 2 diabetes (T2DM) and PD. T2DM is characterized by insulin resistance and failure of β-cells to compensate, leading to hyperglycemia and serious comorbidities. Both PD and T2DM share misregulated processes, including mitochondrial dysfunction, oxidative stress, chronic inflammation, altered proteostasis, protein aggregation, and misregulation of glucose metabolism. Chronic or recurring hyperglycemia is a T2DM hallmark and can lead to increased methylglyoxal (MGO) production, which is responsible for protein glycation. Glycation of alpha-synuclein (aSyn), a central player in PD pathogenesis, accelerates the deleterious aSyn effects. Interestingly, MGO blood plasma levels and aSyn glycation are significantly elevated in T2DM patients, suggesting a molecular mechanism underlying the T2DM - PD link. Compared to high constant glucose levels, glycemic variability (fluctuations in blood glucose levels), can be more detrimental for diabetic patients, causing oxidative stress, inflammation, and endothelial damage. Accordingly, it is imperative for future research to prioritize the exploration of glucose variability's influence on PD development and progression. This involves moving beyond the binary classification of patients as diabetic or non-diabetic, aiming to pave the way for the development of enhanced therapeutic interventions.
Collapse
Affiliation(s)
- Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Science, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
| |
Collapse
|
11
|
Trenkwalder C, Mollenhauer B. The long road to neuroprotection for Parkinson's disease. Lancet Neurol 2024; 23:2-3. [PMID: 38101888 DOI: 10.1016/s1474-4422(23)00462-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023]
Affiliation(s)
- Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany; Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany.
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany; Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Lee HJ, Han K, Kim YW, Yang SN, Yoon SY. Association between lipid levels and the risk of Parkinson's disease in individuals with diabetes mellitus: A nationwide population-based cohort study. Parkinsonism Relat Disord 2023; 117:105881. [PMID: 37951145 DOI: 10.1016/j.parkreldis.2023.105881] [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: 06/29/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 11/13/2023]
Abstract
INTRODUCTION Many studies have examined the positive association between diabetes mellitus (DM) and the risk of Parkinson's disease (PD). Dyslipidemia has been reported to be prevalent in patients with diabetes; thus, lipid levels and the drugs for dyslipidemia could influence the development of PD in patients with DM. This study aimed to examine the association between lipid levels and the risk of PD in individuals with DM and evaluate whether the association changes with the use of statins. METHODS This nationwide population-based retrospective cohort study included individuals with DM according to the International Classification of Diseases between 2009 and 2012. Among the 2,361,633 patients with DM followed up for up to 9 years, 17,046 were newly diagnosed with PD. Patients with DM were categorized into quartile groups of total cholesterol, low-density lipoprotein cholesterol, and triglyceride levels. RESULTS There was an inverse association between lipid levels and PD development in the unadjusted model; however, this relationship became less significant after adjusting the use of statins in triglyceride and total cholesterol. In the analysis stratified by statin use, total cholesterol level was associated with decreased PD risk in non-statin users with DM; however, there was no significant association between total cholesterol level and PD risk in statin users. CONCLUSION We found an inverse relationship between lipid levels and PD risk in patients with DM, which was influenced by statin use. Future studies about optimal target lipid levels relevant to PD risk considering statin dose in DM patients are needed.
Collapse
Affiliation(s)
- Hyo Jeong Lee
- Department of Rehabilitation Medicine, Bundang Jesaeng General Hospital, Seongnam-si, Republic of Korea
| | - Kyungdo Han
- Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Yong Wook Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Nam Yang
- Department of Physical Medicine & Rehabilitation, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Seo Yeon Yoon
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Ogaki K, Fujita H, Nozawa N, Shiina T, Sakuramoto H, Suzuki K. Impact of diabetes and glycated hemoglobin level on the clinical manifestations of Parkinson's disease. J Neurol Sci 2023; 454:120851. [PMID: 37931442 DOI: 10.1016/j.jns.2023.120851] [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: 08/03/2023] [Revised: 09/21/2023] [Accepted: 10/22/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND The coexistence of diabetes mellitus (DM) has been suggested to accelerate the progression of Parkinson's disease (PD) and make the phenotype more severe. In this study, we investigated whether DM or glycated hemoglobin (HbA1c) levels affect the differences in motor and nonmotor symptoms. METHODS We conducted a cross-sectional study including 140 consecutive Japanese patients with PD for whom medical history and serum HbA1c records were available. The PD patients with a DM diagnosis were classified into the diabetes-complicated group (PD-DM) and the nondiabetes-complicated group (PD-no DM). Next, patients were classified based on a median HbA1c value of 5.7, and clinical parameters were compared. The correlations between HbA1c levels and other clinical variables were analyzed. RESULTS Of 140 patients, 23 patients (16%) had DM. Compared to PD-no DM patients, PD-DM patients showed lower MMSE scores. Compared to the lower HbA1c group, the higher HbA1c group showed a higher MDS-UPDRS part III score and a lower metaiodobenzylguanidine (MIBG) scintigraphy heart-to-mediastinum (H/M) ratio. HbA1c levels were positively correlated with age and the MDS-UPDRS part III score and negatively correlated with the MMSE score and H/M ratio on cardiac MIBG scintigraphy. Binary logistic regression analysis, which included age, sex, disease duration, and MMSE and MDS-UPDRS part III scores as independent variables, revealed that a lower MMSE score was an independent contributor to PD-DM and PD with high HbA1c levels. CONCLUSIONS DM complications and high HbA1c levels may affect cognitive function in patients with PD.
Collapse
Affiliation(s)
- Keitaro Ogaki
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Hiroaki Fujita
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan.
| | - Narihiro Nozawa
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Tomohiko Shiina
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | | | - Keisuke Suzuki
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
14
|
Wagner SK, Romero-Bascones D, Cortina-Borja M, Williamson DJ, Struyven RR, Zhou Y, Patel S, Weil RS, Antoniades CA, Topol EJ, Korot E, Foster PJ, Balaskas K, Ayala U, Barrenechea M, Gabilondo I, Schapira AHV, Khawaja AP, Patel PJ, Rahi JS, Denniston AK, Petzold A, Keane PA. Retinal Optical Coherence Tomography Features Associated With Incident and Prevalent Parkinson Disease. Neurology 2023; 101:e1581-e1593. [PMID: 37604659 PMCID: PMC10585674 DOI: 10.1212/wnl.0000000000207727] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/14/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Cadaveric studies have shown disease-related neurodegeneration and other morphological abnormalities in the retina of individuals with Parkinson disease (PD); however, it remains unclear whether this can be reliably detected with in vivo imaging. We investigated inner retinal anatomy, measured using optical coherence tomography (OCT), in prevalent PD and subsequently assessed the association of these markers with the development of PD using a prospective research cohort. METHODS This cross-sectional analysis used data from 2 studies. For the detection of retinal markers in prevalent PD, we used data from AlzEye, a retrospective cohort of 154,830 patients aged 40 years and older attending secondary care ophthalmic hospitals in London, United Kingdom, between 2008 and 2018. For the evaluation of retinal markers in incident PD, we used data from UK Biobank, a prospective population-based cohort where 67,311 volunteers aged 40-69 years were recruited between 2006 and 2010 and underwent retinal imaging. Macular retinal nerve fiber layer (mRNFL), ganglion cell-inner plexiform layer (GCIPL), and inner nuclear layer (INL) thicknesses were extracted from fovea-centered OCT. Linear mixed-effects models were fitted to examine the association between prevalent PD and retinal thicknesses. Hazard ratios for the association between time to PD diagnosis and retinal thicknesses were estimated using frailty models. RESULTS Within the AlzEye cohort, there were 700 individuals with prevalent PD and 105,770 controls (mean age 65.5 ± 13.5 years, 51.7% female). Individuals with prevalent PD had thinner GCIPL (-2.12 μm, 95% CI -3.17 to -1.07, p = 8.2 × 10-5) and INL (-0.99 μm, 95% CI -1.52 to -0.47, p = 2.1 × 10-4). The UK Biobank included 50,405 participants (mean age 56.1 ± 8.2 years, 54.7% female), of whom 53 developed PD at a mean of 2,653 ± 851 days. Thinner GCIPL (hazard ratio [HR] 0.62 per SD increase, 95% CI 0.46-0.84, p = 0.002) and thinner INL (HR 0.70, 95% CI 0.51-0.96, p = 0.026) were also associated with incident PD. DISCUSSION Individuals with PD have reduced thickness of the INL and GCIPL of the retina. Involvement of these layers several years before clinical presentation highlight a potential role for retinal imaging for at-risk stratification of PD.
Collapse
Affiliation(s)
- Siegfried Karl Wagner
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom.
| | - David Romero-Bascones
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Mario Cortina-Borja
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Dominic J Williamson
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Robbert R Struyven
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Yukun Zhou
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Salil Patel
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Rimona S Weil
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Chrystalina A Antoniades
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Eric J Topol
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Edward Korot
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Paul J Foster
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Konstantinos Balaskas
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Unai Ayala
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Maitane Barrenechea
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Iñigo Gabilondo
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Anthony H V Schapira
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Anthony P Khawaja
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Praveen J Patel
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Jugnoo S Rahi
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Alastair K Denniston
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Axel Petzold
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| | - Pearse Andrew Keane
- From the Institute of Ophthalmology (S.K.W., D.J.W., R.R.S., Y.Z., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.P., P.A.K.), University College London; NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology (S.K.W., D.R.-B., D.J.W., R.R.S., Y.Z., E.K., P.J.F., K.B., A.P.K., P.J.P., J.S.R., A.K.D., A.P., P.A.K.), London, United Kingdom; Biomedical Engineering Department (D.R.-B., E.K., U.A., M.B.), Faculty of Engineering (MU-ENG), Mondragon Unibertsitatea, Spain; Great Ormond Street Institute of Child Health (M.C.-B., J.S.R.), and Centre for Medical Image Computing (D.J.W., R.R.S., Y.Z.), Department of Computer Science, University College London; NeuroMetrology Lab (S.P., C.A.A.), Nuffield Department of Clinical Neurosciences, University of Oxford; Dementia Research Centre (R.S.W.), University College London, United Kingdom; Department of Molecular Medicine (E.J.T.), Scripps Research, La Jolla, CA; Byers Eye Institute (E.K.), Stanford University, Palo Alto, CA; Biocruces Bizkaia Health Research Institute (I.G.), Barakaldo; IKERBASQUE: The Basque Foundation for Science (I.G.), Bilbao, Spain; Department of Clinical and Movement Neurosciences (A.H.V.S.), UCL Queen Square Institute of Neurology; Great Ormond Street Hospital NHS Foundation Trust (J.S.R.); Ulverscroft Vision Research Group (J.S.R.), University College London; NIHR Biomedical Research Centre at UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital (J.S.R.), London; University of Birmingham (A.K.D.); University Hospitals Birmingham NHS Foundation Trust (A.K.D.); NIHR Birmingham Biomedical Research Centre (A.K.D.), University of Birmingham; and Queen Square Institute of Neurology (A.P.), University College London, United Kingdom
| |
Collapse
|
15
|
Chuproski AP, Azevedo EM, Ilkiw J, Miloch J, Lima MMS. Metabolic dysfunctions in the intranigral rotenone model of Parkinson's disease. Exp Brain Res 2023; 241:1289-1298. [PMID: 37000202 DOI: 10.1007/s00221-023-06605-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/24/2023] [Indexed: 04/01/2023]
Abstract
Parkinson disease (PD) is a chronic neurodegenerative disorder characterized by a progressive loss of dopamine neurons in the substantia nigra pars compacta (SNpc). In the last years, a growing interest to study the relationship between metabolic dysfunction and neurodegenerative disease like PD has emerged. This study aimed to evaluate the occurrence of possible changes in metabolic homeostasis due to intranigral rotenone administration, a neurotoxin that damages dopaminergic neurons leading to motor impairments mimicking those that happen in PD. Male Wistar rats were distributed into two groups: sham (n = 10) or rotenone (n = 10). Sham group received, bilaterally, within the SNpc, 1 µL of vehicle dimethyl-sulfoxide (DMSO) and the experimental group was bilaterally injected with 1 µL of rotenone (12 µg/µL). Twenty-four hours after the stereotaxic surgeries, the animals underwent the open field test followed by subsequent peripheral blood and cerebrospinal fluid (CSF) samples collection for biochemical testing. The results showed that rotenone was able to replicate the typical motor behavior impairment seen in the disease, i.e., decrease in locomotion (P = 0.05) and increase in immobility (P = 0.01) with a strong correlation (r = - 0.85; P < 0.0001) between them. In addition, it was demonstrated that this model is able to decrease plasmatic total-cholesterol (P = 0.04) and HDL-cholesterol (P = 0.007) potentially impacting peripheral metabolism. Hence, it was revealed a potential ability to reproduce relevant metabolic dysfunctions like hyperglycemia which could be explained by acute and systemic mitochondrial rotenone toxicity and SNpc nigral toxicity. Such mechanisms may still be responsible for the potential occurrence of CSF-hyperglycemia (d = 0.7). Since intranigral rotenone is an early phase model of PD, the present results open a new road for studies aiming to investigate metabolic changes in PD.
Collapse
Affiliation(s)
- Ana Paula Chuproski
- Neurophysiology Laboratory, Department of Physiology, Federal University of Paraná, Setor de Ciências Biológicas, Av. Francisco H. dos Santos s/n, Zip 81.531-990, Curitiba, Paraná, 19031, Brazil
| | - Evellyn Mayla Azevedo
- Neurophysiology Laboratory, Department of Physiology, Federal University of Paraná, Setor de Ciências Biológicas, Av. Francisco H. dos Santos s/n, Zip 81.531-990, Curitiba, Paraná, 19031, Brazil
| | - Jéssica Ilkiw
- Neurophysiology Laboratory, Department of Physiology, Federal University of Paraná, Setor de Ciências Biológicas, Av. Francisco H. dos Santos s/n, Zip 81.531-990, Curitiba, Paraná, 19031, Brazil
| | - Jéssica Miloch
- Neurophysiology Laboratory, Department of Physiology, Federal University of Paraná, Setor de Ciências Biológicas, Av. Francisco H. dos Santos s/n, Zip 81.531-990, Curitiba, Paraná, 19031, Brazil
| | - Marcelo M S Lima
- Neurophysiology Laboratory, Department of Physiology, Federal University of Paraná, Setor de Ciências Biológicas, Av. Francisco H. dos Santos s/n, Zip 81.531-990, Curitiba, Paraná, 19031, Brazil.
- Department of Pharmacology, Federal University of Paraná, Curitiba, Paraná, Brazil.
| |
Collapse
|
16
|
Zhong Q, Wang S. Association between diabetes mellitus, prediabetes and risk, disease progression of Parkinson's disease: A systematic review and meta-analysis. Front Aging Neurosci 2023; 15:1109914. [PMID: 37009459 PMCID: PMC10060805 DOI: 10.3389/fnagi.2023.1109914] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023] Open
Abstract
Background Previous studies reported inconsistent results regarding association between diabetes mellitus (DM), prediabetes and risk, disease progression of Parkinson's disease (PD). The meta-analysis was made to investigate association between DM, prediabetes and risk, disease progression of PD. Methods Literatures investigating association between DM, prediabetes and risk, disease progression of PD were searched in these databases: PubMed and Web of Science. Included literatures were published before October 2022. STATA 12.0 software was used to compute odds ratios (ORs)/relative risks (RRs) or standard mean differences (SMDs). Results DM was associated with a higher risk of PD, compared to non-diabetic participants with a random effects model (OR/RR = 1.23, 95% CI 1.12-1.35, I 2 = 90.4%, p < 0.001). PD with DM (PD-DM) was associated with a faster motor progression compared to PD without DM (PD-noDM) with a fixed effects model (RR = 1.85, 95% CI 1.47-2.34, I 2 = 47.3%, p = 0.091). However, meta-analysis for comparison in change rate of United Rating Scale (UPDRS) III scores from baseline to follow-up time between PD-DM and PD-noDM reported no difference in motor progression between PD-DM and PD-noDM with a random effects model (SMD = 2.58, 95% CI = -3.11 to 8.27, I 2 = 99.9%, p < 0.001). PD-DM was associated with a faster cognitive decline compared to PD-noDM with a fixed effects model (OR/RR = 1.92, 95% CI 1.45-2.55, I 2 = 50.3%, p = 0.110). Conclusions In conclusion, DM was associated with a higher risk and faster disease decline of PD. More large-scale cohort studies should be adopted to evaluate the association between DM, prediabetes and PD.
Collapse
Affiliation(s)
| | - Shenglong Wang
- Department of Neurology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
17
|
Actions and Consequences of Insulin in the Striatum. Biomolecules 2023; 13:biom13030518. [PMID: 36979453 PMCID: PMC10046598 DOI: 10.3390/biom13030518] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
Insulin crosses the blood–brain barrier to enter the brain from the periphery. In the brain, insulin has well-established actions in the hypothalamus, as well as at the level of mesolimbic dopamine neurons in the midbrain. Notably, insulin also acts in the striatum, which shows abundant expression of insulin receptors (InsRs) throughout. These receptors are found on interneurons and striatal projections neurons, as well as on glial cells and dopamine axons. A striking functional consequence of insulin elevation in the striatum is promoting an increase in stimulated dopamine release. This boosting of dopamine release involves InsRs on cholinergic interneurons, and requires activation of nicotinic acetylcholine receptors on dopamine axons. Opposing this dopamine-enhancing effect, insulin also increases dopamine uptake through the action of insulin at InsRs on dopamine axons. Insulin acts on other striatal cells as well, including striatal projection neurons and astrocytes that also influence dopaminergic transmission and striatal function. Linking these cellular findings to behavior, striatal insulin signaling is required for the development of flavor–nutrient learning, implicating insulin as a reward signal in the brain. In this review, we discuss these and other actions of insulin in the striatum, including how they are influenced by diet and other physio-logical states.
Collapse
|
18
|
Amagai R, Yoshioka S, Otomo R, Nagano H, Hashimoto N, Sakakibara R, Tanaka T, Okado-Matsumoto A. Post-translational modification of lysine residues in erythrocyte α-synuclein. J Biochem 2023; 173:177-184. [PMID: 36469357 DOI: 10.1093/jb/mvac100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/30/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
α-Synuclein is a protein linked to various synuclein-associated diseases ('synucleinopathies'), including Parkinson's disease, dementia with Lewy Bodies and multiple system atrophy, and is highly expressed in the central nervous system and in erythrocytes. Moreover, α-synuclein-containing erythrocyte-derived extracellular vesicles may be involved in the pathogenesis of synucleinopathies and their progression across the blood-brain barrier. Several post-translational modifications of α-synuclein have been reported in brain inclusions, including S129 phosphorylation, but fewer have been found in erythrocytes. In this study, we analysed the post-translational modifications of erythrocyte α-synuclein using liquid chromatography-mass spectrometry. We found that all lysine residues in the α-synuclein protein could be modified by acetylation, glycation, ubiquitination or SUMOylation but that phosphorylation, nitration and acylation were uncommon minor post-translational modifications in erythrocytes. Since the post-translational modification of lysine residues has been implicated in both membrane association and protein clearance, our findings provide new insight into how synucleinopathies may progress and suggest possible therapeutic strategies designed to target α-synuclein.
Collapse
Key Words
- Parkinson’s disease
- erythrocyte
- post-translational modification
- synucleinopathy
- α-synuclein.Abbreviations: aa, amino acids; AGE, advanced glycation end product; BBB, blood–brain barrier; CML, Nε-(1-carboxymethyl)-L-lysine; CNS, central nervous system; EVs, extracellular vesicles; IP, immunoprecipitation; LC–MS/MS, liquid chromatography–mass spectrometry; PBS, phosphate buffered saline; PD, Parkinson’s disease; PTM, post-translational modification; SUMO, small ubiquitin-related modifier
Collapse
Affiliation(s)
- Ryosuke Amagai
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Sakura Yoshioka
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Riki Otomo
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Hidekazu Nagano
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Naoko Hashimoto
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ryuji Sakakibara
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Chiba 285-8741, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Chiba University Graduate School of Medicine, Chiba, Chiba 260-8670, Japan
| | - Ayako Okado-Matsumoto
- Laboratory of Biochemistry, Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
19
|
Selim R, Gordon SC, Zhou Y, Zhang T, Lu M, Daida YG, Boscarino JA, Schmidt MA, Trudeau S, Rupp LB, Gonzalez HC. Impact of hepatitis C treatment status on risk of Parkinson's disease and secondary parkinsonism in the era of direct-acting antivirals. J Viral Hepat 2023; 30:544-550. [PMID: 36872452 DOI: 10.1111/jvh.13826] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/10/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Research suggests a possible link between chronic infection with hepatitis C virus (HCV) and the development of Parkinson's Disease (PD) and secondary Parkinsonism (PKM). We investigated the impact of antiviral treatment status (untreated, interferon [IFN] treated, direct-acting antiviral [DAA] treated) and outcome (treatment failure [TF] or sustained virological response [SVR]) on risk of PD/PKM among patients with HCV. Using data from the Chronic Hepatitis Cohort Study (CHeCS), we applied a discrete time-to-event approach with PD/PKM as the outcome. We performed univariate followed by a multivariable modelling that used time-varying covariates, propensity scores to adjust for potential treatment selection bias and death as a competing risk. Among 17,199 confirmed HCV patients, we observed 54 incident cases of PD/PKM during a mean follow-up period of 17 years; 3753 patients died during follow-up. There was no significant association between treatment status/outcome and risk of PD/PKM. Type 2 diabetes tripled risk (hazard ratio [HR] 3.05; 95% CI 1.75-5.32; p < .0001) and presence of cirrhosis doubled risk of PD/PKM (HR 2.13, 95% CI 1.31-3.47). BMI >30 was associated with roughly 50% lower risk of PD/PKM than BMI <25 (HR 0.43; 0.22-0.84; p = .0138). After adjustment for treatment selection bias, we did not observe a significant association between HCV patients' antiviral treatment status/outcome on risk of PD/PKM. Several clinical risk factors-diabetes, cirrhosis and BMI-were associated with PD/PKM.
Collapse
Affiliation(s)
- Ranya Selim
- Department of Gastroenterology and Hepatology, Henry Ford Health, Detroit, Michigan, United States
| | - Stuart C Gordon
- Department of Gastroenterology and Hepatology, Henry Ford Health, Detroit, Michigan, United States.,School of Medicine, Wayne State University, Detroit, Michigan, United States
| | - Yueren Zhou
- Department of Public Health Sciences, Henry Ford Health, Detroit, Michigan, United States
| | - Talan Zhang
- Center on Aging & Health, Johns Hopkins University, Baltimore, Maryland, United States
| | - Mei Lu
- Center on Aging & Health, Johns Hopkins University, Baltimore, Maryland, United States
| | - Yihe G Daida
- Center for Integrated Health Care Research, Kaiser Permanente Hawaii, Honolulu, Hawaii, United States
| | | | - Mark A Schmidt
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon, United States
| | - Sheri Trudeau
- Center on Aging & Health, Johns Hopkins University, Baltimore, Maryland, United States
| | - Loralee B Rupp
- Department of Health Policy and Health Systems Research, Henry Ford Health, Detroit, Michigan, United States
| | - Humberto C Gonzalez
- Department of Gastroenterology and Hepatology, Henry Ford Health, Detroit, Michigan, United States.,School of Medicine, Wayne State University, Detroit, Michigan, United States
| |
Collapse
|
20
|
Cullinane PW, de Pablo Fernandez E, König A, Outeiro TF, Jaunmuktane Z, Warner TT. Type 2 Diabetes and Parkinson's Disease: A Focused Review of Current Concepts. Mov Disord 2023; 38:162-177. [PMID: 36567671 DOI: 10.1002/mds.29298] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 12/27/2022] Open
Abstract
Highly reproducible epidemiological evidence shows that type 2 diabetes (T2D) increases the risk and rate of progression of Parkinson's disease (PD), and crucially, the repurposing of certain antidiabetic medications for the treatment of PD has shown early promise in clinical trials, suggesting that the effects of T2D on PD pathogenesis may be modifiable. The high prevalence of T2D means that a significant proportion of patients with PD may benefit from personalized antidiabetic treatment approaches that also confer neuroprotective benefits. Therefore, there is an immediate need to better understand the mechanistic relation between these conditions and the specific molecular pathways affected by T2D in the brain. Although there is considerable evidence that processes such as insulin signaling, mitochondrial function, autophagy, and inflammation are involved in the pathogenesis of both PD and T2D, the primary aim of this review is to highlight the evidence showing that T2D-associated dysregulation of these pathways occurs not only in the periphery but also in the brain and how this may facilitate neurodegeneration in PD. We also discuss the challenges involved in disentangling the complex relationship between T2D, insulin resistance, and PD, as well as important questions for further research. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Patrick W Cullinane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Eduardo de Pablo Fernandez
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Annekatrin König
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom.,Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London NHS Foundation Trust, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.,Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,Queen Square Movement Disorders Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
21
|
Park J, Choi S, Kim R. Association between prediabetes and cognitive function in Parkinson's disease. Brain Behav 2023; 13:e2838. [PMID: 36448303 PMCID: PMC9847602 DOI: 10.1002/brb3.2838] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION It remains largely unknown whether prediabetes is related to cognitive impairment in Parkinson's disease (PD). This study aimed to assess the association between prediabetes and cognitive function in PD patients. METHODS In this cross-sectional study, 262 PD patients (age, 69.8 ± 10.3 years; Hoehn-Yahr stage, 2.3 ± 0.8) were classified into diabetes (glycated hemoglobin [HbA1c] ≥6.5% or previously diagnosed, n = 76), prediabetes (5.7%-6.4%, n = 90), or diabetes free (≤5.6%, n = 96) groups. Cognitive function was measured using the Montreal Cognitive Assessment (MoCA) test. RESULTS Both the diabetes and prediabetes groups had significantly lower MoCA scores (17.0 ± 6.6 and 18.0 ± 6.1, respectively) than the diabetes free group (20.0 ± 5.7), even after adjusting for potential confounders (p = .002 and p = .008, respectively). In the combined group of prediabetes and diabetes free patients, higher HbA1c levels significantly correlated with lower MoCA scores (p = .031). There was a significant interaction of diabetes status with age, but not with the duration of PD, on cognitive function. CONCLUSION In addition to diabetes, prediabetes may negatively affect cognitive function in PD patients. Further prospective longitudinal studies are necessary to clarify the impact of prediabetes on the cognitive trajectory of these patients.
Collapse
Affiliation(s)
- Joah Park
- Department of Medicine, Inha University College of Medicine, Incheon, Korea
| | - Seohee Choi
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Ryul Kim
- Department of Neurology, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
22
|
Yu H, Sun T, He X, Wang Z, Zhao K, An J, Wen L, Li JY, Li W, Feng J. Association between Parkinson's Disease and Diabetes Mellitus: From Epidemiology, Pathophysiology and Prevention to Treatment. Aging Dis 2022; 13:1591-1605. [PMID: 36465171 PMCID: PMC9662283 DOI: 10.14336/ad.2022.0325] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/25/2022] [Indexed: 08/27/2023] Open
Abstract
Diabetes mellitus (DM) and Parkinson's disease (PD) are both age-related diseases of global concern being among the most common chronic metabolic and neurodegenerative diseases, respectively. While both diseases can be genetically inherited, environmental factors play a vital role in their pathogenesis. Moreover, DM and PD have common underlying molecular mechanisms, such as misfolded protein aggregation, mitochondrial dysfunction, oxidative stress, chronic inflammation, and microbial dysbiosis. Recently, epidemiological and experimental studies have reported that DM affects the incidence and progression of PD. Moreover, certain antidiabetic drugs have been proven to decrease the risk of PD and delay its progression. In this review, we elucidate the epidemiological and pathophysiological association between DM and PD and summarize the antidiabetic drugs used in animal models and clinical trials of PD, which may provide reference for the clinical translation of antidiabetic drugs in PD treatment.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhen Wang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
| | - Kaidong Zhao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
| | - Jing An
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Lulu Wen
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jia-Yi Li
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Wen Li
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, Shenyang, Liaoning, China.
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
23
|
Ferrari F, Moretti A, Villa RF. Incretin-based drugs as potential therapy for neurodegenerative diseases: current status and perspectives. Pharmacol Ther 2022; 239:108277. [DOI: 10.1016/j.pharmthera.2022.108277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
|
24
|
Sun X, Xue L, Wang Z, Xie A. Update to the Treatment of Parkinson's Disease Based on the Gut-Brain Axis Mechanism. Front Neurosci 2022; 16:878239. [PMID: 35873830 PMCID: PMC9299103 DOI: 10.3389/fnins.2022.878239] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/20/2022] [Indexed: 12/27/2022] Open
Abstract
Gastrointestinal (GI) symptoms represented by constipation were significant non-motor symptoms of Parkinson’s disease (PD) and were considered early manifestations and aggravating factors of the disease. This paper reviewed the research progress of the mechanism of the gut-brain axis (GBA) in PD and discussed the roles of α-synuclein, gut microbiota, immune inflammation, neuroendocrine, mitochondrial autophagy, and environmental toxins in the mechanism of the GBA in PD. Treatment of PD based on the GBA theory has also been discussed, including (1) dietary therapy, such as probiotics, vitamin therapy, Mediterranean diet, and low-calorie diet, (2) exercise therapy, (3) drug therapy, including antibiotics; GI peptides; GI motility agents, and (4) fecal flora transplantation can improve the flora. (5) Vagotomy and appendectomy were associated but not recommended.
Collapse
Affiliation(s)
- Xiaohui Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Xue
- Recording Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zechen Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|