2
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Diabetes and Neuroaxonal Damage in Parkinson's Disease

      letter

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          We read with interest Uyar and colleagues' recent report on the association between diabetes, nondiabetic elevated glycated hemoglobin levels (HbA1c), and neuroaxonal damage in Parkinson's disease (PD) patients from the MARK‐PD study. 1 The authors confirmed previously established findings of an inverse association between diabetes and cognitive and motor status. The authors also demonstrated higher serum neurofilament light (NfL) levels (a marker of neuroaxonal damage) 2 in PD patients with prevalent type 2 diabetes and in PD patients with nondiabetic elevated HbA1c levels. These associations persisted after adjustment for age, body mass index (BMI), and vascular risk factors (prevalent arterial hypertension, hypercholesterolemia, and history of stroke). We recently noted similar motor and cognitive associations in PD patients with diabetes 3 in the Tracking Parkinson's study, although only a nonsignificant trend toward an association in the overall PD cohort between NfL levels and more severe motor and cognitive status at baseline,4 which may reflect the reduced disease duration in the Tracking Parkinson's cohort, compared with the MARK‐PD cohort. Considering the authors' novel findings of an association between diabetes and neuroaxonal damage, we explored the relationship between serum NfL and diabetes in our previously defined subgroup of the Tracking Parkinson's study. 4 The analysis was performed using Stata V.17.0 (Stata, RRID:SCR_012763), and differences were compared using Kruskal–Wallis tests for continuous data and χ2 tests for categorical data, whereas the association between NfL and diabetes was further explored using univariate and multivariate (age, BMI, and vascular risk factors) linear regression analysis. Of the 280 patients studied, 29 suffered from prevalent type 2 diabetes. PD‐DM patients were older (74.1 years ± SD 7.7 vs. 68.1 years ± 8.7, P < 0.001), with higher BMIs (31.1 ± SD [standard deviation] 5.7 vs. 27.1 ± SD 4.4, P < 0.001), whereas a higher proportion had coexistent vascular risk factors than PD patients without diabetes (P = 0.032). Serum NfL levels were higher in PD‐DM patients (39.5 ± SD 18.9 vs. 29.6 ± SD 16.0, P < 0.001). Using regression analysis, NfL levels were significantly associated with patients' diabetic status (coefficient: 0.82, 95% CI [confidence interval]: 0.45–1.19, P < 0.0001), which persisted (coefficient: 0.52, 95% CI: 0.18–0.86, P = 0.003) after adjustment for age, BMI, and vascular risk factors (history of angina, myocardial infarction, stroke, hypertension, and hypercholesterolemia). Our findings affirm Uyar et al's report of an association between PD‐DM and more severe neuroaxonal damage. Furthermore, the data indicate that the more severe phenotype in PD‐DM noted to date by several studies is likely to be mediated by additional factors other than vascular risk factor burden that tends to coexist in these cases. T2DM and PD share several pathological processes encompassing neuroinflammation, lysosomal dysfunction, mitochondrial dysfunction, and the development of central insulin resistance that leads to neurodegeneration. 5 This process is in part mediated by hyperglycemia as demonstrated by the MARK‐PD study and its downstream impact on α‐synuclein aggregation. 6 It is also possible that some of the observed associations are explained by diabetic neuropathy, as other peripheral neuropathies are known to increase blood NfL concentrations. 7 Disentangling the mechanistic factors that contribute to this more rapidly progressive axonal damage is of critical importance in the development of disease‐modifying therapies for PD. Full financial disclosures for the previous 12 months N.V. has received unconditional educational grants from Ipsen and Biogen; travel grants from Ipsen, AbbVie, and the International Parkinson's Disease and Movement Disorders Society; and speaker's honorarium from AbbVie and Stada and served on advisory boards for AbbVie and Brittania outside of the submitted work. M.L. has no competing interest. R.R. has no competing interest. A.J.H. has no competing interest. T.G. has no competing interest. D.A. has no competing interest. C.G. has no competing interest. Y.B.‐S. has no competing interest. H.Z. has served at scientific advisory boards for AbbVie, Alector, Eisai, Denali, Roche Diagnostics, Wave, Samumed, Siemens Healthineers, Pinteon Therapeutics, Nervgen, AZTherapies, and CogRx; has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure, and Biogen; and is a cofounder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside the submitted work). D.G.G. has received honoraria from Bial Pharma, GE Healthcare, and Vectura plc and consultancy fees from the Glasgow Memory Clinic. H.R.M. is employed by UCL. In the past 24 months he reports paid consultancy from Biogen, UCB, AbbVie, Denali, Biohaven, and Lundbeck; lecture fees/honoraria from Biogen, UCB, C4X Discovery, GE‐Healthcare, Wellcome Trust, and Movement Disorders Society; and research grants from ASAP, Parkinson's UK, Cure Parkinson's Trust, PSP Association, CBD Solutions, Drake Foundation, and Medical Research Council. Dr. Morris is a co‐applicant on a patent application related to C9ORF72—Method for diagnosing a neurodegenerative disease (PCT/GB2012/052140). T.F. has received grants from the National Institute of Health Research, The Michael J. Fox Foundation, John Black Charitable Foundation, Cure Parkinson's Trust, Innovate UK, Van Andel Research Institute, and Defeat MSA. He has served on advisory boards for Voyager Therapeutics, Handl Therapeutics, Living Cell Technologies, Bial, and Profile Pharma. He has received honoraria for talks sponsored by Bial, Profile Pharma, and Boston Scientific. Author Roles Research project: A. Conception, B. Organization, C. Execution; Manuscript preparation: A. Writing of the first draft, B. Review and critique. N.V.: 1A, 1B, 1C, 2A, 2B M.L.: 1C, 2B R.R.: 1C, 2B A.J.H.: 1C, 2B T.G.: 1C, 2B D.A.: 1C, 2B C.G.: 1C, 2B Y.B.‐S.: 1C, 2B H.Z.: 1C, 2B D.G.G.: 1A, 1B, 1C, 2B H.R.M.: 1A, 1B, 1C, 2B T.F.: 1A, 1B, 1C, 2B.

          Related collections

          Most cited references8

          • Record: found
          • Abstract: found
          • Article: found

          Neurofilament light chain as a biomarker in neurological disorders

          In the management of neurological diseases, the identification and quantification of axonal damage could allow for the improvement of diagnostic accuracy and prognostic assessment. Neurofilament light chain (NfL) is a neuronal cytoplasmic protein highly expressed in large calibre myelinated axons. Its levels increase in cerebrospinal fluid (CSF) and blood proportionally to the degree of axonal damage in a variety of neurological disorders, including inflammatory, neurodegenerative, traumatic and cerebrovascular diseases. New immunoassays able to detect biomarkers at ultralow levels have allowed for the measurement of NfL in blood, thus making it possible to easily and repeatedly measure NfL for monitoring diseases’ courses. Evidence that both CSF and blood NfL may serve as diagnostic, prognostic and monitoring biomarkers in neurological diseases is progressively increasing, and NfL is one of the most promising biomarkers to be used in clinical and research setting in the next future. Here we review the most important results on CSF and blood NfL and we discuss its potential applications and future directions.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Insulin resistance and Parkinson's disease: A new target for disease modification?

            There is growing evidence that patients with Type 2 diabetes have an increased risk of developing Parkinson's disease and share similar dysregulated pathways suggesting common underlying pathological mechanisms. Historically insulin was thought solely to be a peripherally acting hormone responsible for glucose homeostasis and energy metabolism. However accumulating evidence indicates insulin can cross the blood-brain-barrier and influence a multitude of processes in the brain including regulating neuronal survival and growth, dopaminergic transmission, maintenance of synapses and pathways involved in cognition. In conjunction, there is growing evidence that a process analogous to peripheral insulin resistance occurs in the brains of Parkinson's disease patients, even in those without diabetes. This raises the possibility that defective insulin signalling pathways may contribute to the development of the pathological features of Parkinson's disease, and thereby suggests that the insulin signalling pathway may potentially be a novel target for disease modification. Given these growing links between PD and Type 2 diabetes it is perhaps not unsurprising that drugs used the treatment of T2DM are amongst the most promising treatments currently being prioritised for repositioning as possible novel treatments for PD and several clinical trials are under way. In this review, we will examine the underlying cellular links between insulin resistance and the pathogenesis of PD and then we will assess current and future pharmacological strategies being developed to restore neuronal insulin signalling as a potential strategy for slowing neurodegeneration in Parkinson's disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Glycation potentiates α-synuclein-associated neurodegeneration in synucleinopathies.

              α-Synuclein misfolding and aggregation is a hallmark in Parkinson's disease and in several other neurodegenerative diseases known as synucleinopathies. The toxic properties of α-synuclein are conserved from yeast to man, but the precise underpinnings of the cellular pathologies associated are still elusive, complicating the development of effective therapeutic strategies. Combining molecular genetics with target-based approaches, we established that glycation, an unavoidable age-associated post-translational modification, enhanced α-synuclein toxicity in vitro and in vivo, in Drosophila and in mice. Glycation affected primarily the N-terminal region of α-synuclein, reducing membrane binding, impaired the clearance of α-synuclein, and promoted the accumulation of toxic oligomers that impaired neuronal synaptic transmission. Strikingly, using glycation inhibitors, we demonstrated that normal clearance of α-synuclein was re-established, aggregation was reduced, and motor phenotypes in Drosophila were alleviated. Altogether, our study demonstrates glycation constitutes a novel drug target that can be explored in synucleinopathies as well as in other neurodegenerative conditions.
                Bookmark

                Author and article information

                Contributors
                t.foltynie@ucl.ac.uk
                Journal
                Mov Disord
                Mov Disord
                10.1002/(ISSN)1531-8257
                MDS
                Movement Disorders
                John Wiley & Sons, Inc. (Hoboken, USA )
                0885-3185
                1531-8257
                20 July 2022
                July 2022
                : 37
                : 7 ( doiID: 10.1002/mds.v37.7 )
                : 1568-1569
                Affiliations
                [ 1 ] Department of Clinical and Movement Neurosciences UCL Queen Square Institute of Neurology London United Kingdom
                [ 2 ] School of Social and Community Medicine University of Bristol Bristol United Kingdom
                [ 3 ] Department of Social Medicine University of Bristol Bristol United Kingdom
                [ 4 ] Aligning Science Across Parkinson's (ASAP) Collaborative Research Network Chevy Chase Maryland USA
                [ 5 ] Dementia Research Institute University College London London United Kingdom
                [ 6 ] Department of Neurodegenerative Disease UCL Institute of Neurology, Queen, Square London United Kingdom
                [ 7 ] Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Mölndal Sweden
                [ 8 ] Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg Mölndal Sweden
                [ 9 ] Hong Kong Center, for Neurodegenerative Diseases Hong Kong People's Republic of China
                [ 10 ] Department of Neurology, Southern General Hospital University of Glasgow and Institute of Neurological Sciences Glasgow United Kingdom
                Author notes
                [*] [* ] Correspondence to: Prof. Thomas Foltynie, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK; E‐mail: t.foltynie@ 123456ucl.ac.uk

                Author information
                https://orcid.org/0000-0002-9671-0212
                https://orcid.org/0000-0001-8594-2483
                https://orcid.org/0000-0003-0752-1813
                Article
                MDS29067
                10.1002/mds.29067
                9543586
                35856732
                72864fbf-bb2e-4d2d-9e34-2dd1df06b3c9
                © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 12 April 2022
                : 18 April 2022
                Page count
                Figures: 0, Tables: 0, Pages: 12, Words: 1723
                Funding
                Funded by: Aligning Science Across Parkinson's initiative
                Award ID: ASAP‐000478
                Funded by: Alzheimer Drug Discovery Foundation
                Award ID: 201809‐2016862
                Funded by: Centrum för idrottsforskning , doi 10.13039/501100005350;
                Award ID: 2018‐02532
                Funded by: H2020 European Research Council , doi 10.13039/100010663;
                Award ID: 681712
                Award ID: 101053962
                Funded by: Swedish State Support for Clinical Research
                Award ID: ALFGBG‐71320
                Funded by: the AD Strategic Fund and the Alzheimer's Association
                Award ID: ADSF‐21‐831376‐C
                Award ID: #ADSF‐21‐831381‐C
                Award ID: #ADSF‐21‐8
                Funded by: the European Union Joint Programme ‐ Neurodegenerative Disease Research
                Award ID: JPND2021‐00694
                Funded by: the European Union's Horizon 2020 research and innovation programme under the Marie Skłodowska‐Curie grant agreement
                Award ID: 860197
                Funded by: the Olav Thon Foundation, the Erling‐Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden
                Award ID: FO2019‐0228
                Funded by: the UK Dementia Research Institute at UCL
                Award ID: UKDRI‐1003
                Funded by: Parkinson's UK , doi 10.13039/501100000304;
                Categories
                Letters: Published Article
                Regular Issue Articles
                Letters: Published Articles
                Custom metadata
                2.0
                July 2022
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.2.0 mode:remove_FC converted:07.10.2022

                Medicine
                Medicine

                Comments

                Comment on this article