51
|
Xu X, Liao J, Dong Q, Qin F, Li J, Sun X, Lu T, Fang L, Peng F, Lu Z, Qiu W. Clinical utility of SUDOSCAN in predicting autonomic neuropathy in patients with Parkinson's disease. Parkinsonism Relat Disord 2019; 64:60-65. [DOI: 10.1016/j.parkreldis.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 03/03/2019] [Accepted: 03/11/2019] [Indexed: 11/27/2022]
|
52
|
Elevated Serum Homocysteine Levels Have Differential Gender-Specific Associations with Motor and Cognitive States in Parkinson's Disease. PARKINSONS DISEASE 2019; 2019:3124295. [PMID: 31275543 PMCID: PMC6560330 DOI: 10.1155/2019/3124295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022]
Abstract
Background Studies attempting to elucidate an association between homocysteine and symptom progression in Parkinson's disease (PD) have had largely discrepant findings. This study aimed to investigate elevated serum homocysteine levels and symptom progression in a cohort of PD patients. Methods Serum homocysteine, folate, and vitamin B12 levels were measured in 205 people with PD and 78 age-matched healthy controls. People with Parkinson's disease underwent a battery of clinical assessments to evaluate symptom severity, including motor (MDS-UPDRS) and cognitive (ACE-R) assessments. Multivariate generalised linear models were created, controlling for confounding variables, and were used to determine whether serum markers are associated with various symptom outcome measures. Results People with Parkinson's disease displayed significantly elevated homocysteine levels (p < 0.001), but not folate or vitamin B12 levels, when compared to healthy controls. A significant positive correlation between homocysteine and MDS-UPDRS III score was identified in males with Parkinson's disease (rs = 0.319, p < 0.001), but not in females, whereas a significant negative correlation between homocysteine levels and total ACE-R score was observed in females with Parkinson's disease (rs = −0.449, p < 0.001), but not in males. Multivariate general linear models confirmed that homocysteine was significantly predictive of MDS-UPDRS III score in male patients (p=0.004) and predictive of total ACE-R score in female patients (p=0.021). Conclusion Elevated serum homocysteine levels are associated with a greater motor impairment in males with Parkinson's disease and poorer cognitive performance in females with Parkinson's disease. Our gender-specific findings may help to explain previous discrepancies in the literature surrounding the utility of homocysteine as a biomarker in PD.
Collapse
|
53
|
Myelinating Glia-Specific Deletion of Fbxo7 in Mice Triggers Axonal Degeneration in the Central Nervous System Together with Peripheral Neuropathy. J Neurosci 2019; 39:5606-5626. [PMID: 31085610 DOI: 10.1523/jneurosci.3094-18.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/10/2019] [Accepted: 04/24/2019] [Indexed: 12/25/2022] Open
Abstract
Myelination of axons facilitates the rapid propagation of electrical signals and the long-term integrity of axons. The ubiquitin-proteasome system is essential for proper protein homeostasis, which is particularly crucial for interactions of postmitotic cells. In our study, we examined how the E3 ubiquitin ligase FBXO7-SCF (SKP1, Cul1, F-box protein) expressed in myelinating cells affects the axon-myelin unit. Deletion of Fbxo7 in oligodendrocytes and Schwann cells in mice using the Cnp1-Cre driver line led to motor impairment due to hindlimb paresis. It did not result in apoptosis of myelinating cells, nor did it affect the proper myelination of axons or lead to demyelination. It however triggered axonal degeneration in the CNS and resulted in the severe degeneration of axons in the PNS, inducing a full-blown neuropathy. Both the CNS and PNS displayed inflammation, while the PNS was also characterized by fibrosis, massive infiltration of macrophages, and edema. Tamoxifen-induced deletion of Fbxo7, after myelination using the Plp1-CreERT2 line, led to a small number of degenerated axons and hence a very mild peripheral neuropathy. Interestingly, loss of Fbxo7 also resulted in reduced proteasome activity in Schwann cells but not in cerebellar granule neurons, indicating a specific sensitivity of the former cell type. Together, our results demonstrate an essential role for FBXO7 in myelinating cells to support associated axons, which is fundamental to the proper developmental establishment and the long-term integrity of the axon-myelin unit.SIGNIFICANCE STATEMENT The myelination of axons facilitates the fast propagation of electrical signals and the trophic support of the myelin-axon unit. Here, we report that deletion of Fbxo7 in myelinating cells in mice triggered motor impairment but had no effect on myelin biogenesis. Loss of Fbxo7 in myelinating glia, however, led to axonal degeneration in the CNS and peripheral neuropathy of the axonal type. In addition, we found that Schwann cells were particularly sensitive to Fbxo7 deficiency reflected by reduced proteasome activity. Based on these findings, we conclude that Fbxo7 is essential for the support of the axon-myelin unit and long-term axonal health.
Collapse
|
54
|
Schaffner A, Li X, Gomez-Llorente Y, Leandrou E, Memou A, Clemente N, Yao C, Afsari F, Zhi L, Pan N, Morohashi K, Hua X, Zhou MM, Wang C, Zhang H, Chen SG, Elliott CJ, Rideout H, Ubarretxena-Belandia I, Yue Z. Vitamin B 12 modulates Parkinson's disease LRRK2 kinase activity through allosteric regulation and confers neuroprotection. Cell Res 2019; 29:313-329. [PMID: 30858560 PMCID: PMC6462009 DOI: 10.1038/s41422-019-0153-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/09/2019] [Indexed: 12/12/2022] Open
Abstract
Missense mutations in Leucine-Rich Repeat Kinase 2 (LRRK2) cause the majority of familial and some sporadic forms of Parkinson's disease (PD). The hyperactivity of LRRK2 kinase induced by the pathogenic mutations underlies neurotoxicity, promoting the development of LRRK2 kinase inhibitors as therapeutics. Many potent and specific small-molecule LRRK2 inhibitors have been reported with promise. However, nearly all inhibitors are ATP competitive-some with unwanted side effects and unclear clinical outcome-alternative types of LRRK2 inhibitors are lacking. Herein we identify 5'-deoxyadenosylcobalamin (AdoCbl), a physiological form of the essential micronutrient vitamin B12 as a mixed-type allosteric inhibitor of LRRK2 kinase activity. Multiple assays show that AdoCbl directly binds LRRK2, leading to the alterations of protein conformation and ATP binding in LRRK2. STD-NMR analysis of a LRRK2 homologous kinase reveals the contact sites in AdoCbl that interface with the kinase domain. Furthermore, we provide evidence that AdoCbl modulates LRRK2 activity through disrupting LRRK2 dimerization. Treatment with AdoCbl inhibits LRRK2 kinase activity in cultured cells and brain tissue, and prevents neurotoxicity in cultured primary rodent neurons as well as in transgenic C. elegans and D. melanogaster expressing LRRK2 disease variants. Finally, AdoCbl alleviates deficits in dopamine release sustainability caused by LRRK2 disease variants in mouse models. Our study uncovers vitamin B12 as a novel class of LRRK2 kinase modulator with a distinct mechanism, which can be harnessed to develop new LRRK2-based PD therapeutics in the future.
Collapse
Affiliation(s)
- Adam Schaffner
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xianting Li
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yacob Gomez-Llorente
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emmanouela Leandrou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Anna Memou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Nicolina Clemente
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Chen Yao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Farinaz Afsari
- Department of Biology, University of York, York, YO1 5DD, UK
| | - Lianteng Zhi
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Nina Pan
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Keita Morohashi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xiaoluan Hua
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ming-Ming Zhou
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chunyu Wang
- Department of Biological Sciences, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Hui Zhang
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Shu G Chen
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | - Hardy Rideout
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Iban Ubarretxena-Belandia
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Biofisika Institute (CSIC, UPV/EHU), University of the Basque Country, Leioa, Spain
| | - Zhenyu Yue
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
55
|
Jeziorska M, Atkinson A, Kass-Iliyya L, Javed S, Kobylecki C, Gosal D, Marshall A, Silverdale M, Malik RA. Increased Intraepidermal Nerve Fiber Degeneration and Impaired Regeneration Relate to Symptoms and Deficits in Parkinson's Disease. Front Neurol 2019; 10:111. [PMID: 30837937 PMCID: PMC6383044 DOI: 10.3389/fneur.2019.00111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Previous studies have shown cutaneous small fiber pathology in patients with Parkinson's disease (PD). These studies have focused on nerve degeneration, but recent reports suggest that nerve regeneration may also be important in PD pathology. Objective: To establish the extent of intraepidermal nerve fiber (IENF) degeneration and regeneration and its relationship to clinical and neurological deficits in Parkinson's disease (PD). Methods: Twenty-three PD patients and 10 age-matched controls underwent skin biopsy and assessment of somatic and autonomic symptoms and deficits. We have assessed Intraepidermal Nerve Fiber Density (IENFD) using standard PGP9.5 staining and GAP-43 to assess Mean Axonal Length (MAL) and Intraepidermal Total Nerve Fiber Length (IETNFL). Results: IENFD (p < 0.0001), MAL (p < 0.0001), IETNFL/Area (p = 0.009), and IETNFL/Length (p = 0.04) were significantly reduced in patients with PD compared to controls. IENFD correlated significantly with disease duration (p = 0.03), cumulative levodopa dose (p = 0.02), Unified Parkinson's Disease Rating Scale, Part III (UPDRS-III) (p = 0.01), Schwab and England Activities of Daily Living (ADL) (p = 0.03), NSP (p = 0.03), and 30:15 ratio (p = 0.03). IETNFL/Area correlated with the Autonomic Scale for Outcomes in Parkinson's Disease (SCOPA-AUT) (p = 0.03) and Diabetic Neuropathy Symptom score (DNS) (p = 0.04) and IETNFL/Length correlated with DNS (p = 0.03). MAL correlated with SCOPA-AUT (p = 0.01), DNS (p = 0.02), and DB-HRV (p = 0.02). Conclusion: Increased IENF degeneration and impaired regeneration correlates with somatic and autonomic symptoms and deficits in patients with PD.
Collapse
Affiliation(s)
- Maria Jeziorska
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Andrew Atkinson
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Lewis Kass-Iliyya
- Department of Neurology, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom.,Division of Neuroscience and Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Saad Javed
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Christopher Kobylecki
- Department of Neurology, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom.,Division of Neuroscience and Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - David Gosal
- Department of Neurology, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Andrew Marshall
- Division of Neuroscience and Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom.,Department of Clinical Neurophysiology, Central Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Monty Silverdale
- Department of Neurology, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom.,Division of Neuroscience and Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Rayaz A Malik
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom.,Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
56
|
Romagnolo A, Merola A, Artusi CA, Rizzone MG, Zibetti M, Lopiano L. Levodopa-Induced Neuropathy: A Systematic Review. Mov Disord Clin Pract 2018; 6:96-103. [PMID: 30838307 DOI: 10.1002/mdc3.12688] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/30/2022] Open
Abstract
Background Clinical, neurophysiological, and pathological evidence suggest an association between Parkinson's disease (PD) and peripheral neuropathy (PNP), with a possible causative role of levodopa metabolic products, such as homocysteine and methylmalonic acid. Methods We conducted a systematic review of studies reporting cases of PNP in l-dopa-treated PD patients indexed in PubMed between January 1990 and March 2018. Results We identified 38 articles reporting cases of PNP in PD patients treated with oral l-dopa or with l-dopa/carbidopa intestinal gel infusion (LCIG). Prevalence of PNP was 30.2% in the former group and 42.1% in the latter. Oral l-dopa was mostly associated with slowly progressive PNP, whereas LCIG showed an acute or subacute onset in 35.7% of cases. In both groups, there was an association between PNP and higher l-dopa doses, as well as with the following biochemical alterations: increased homocysteine; reduced vitamin B12; increased methylmalonic acid; and reduced vitamin B6. A skin biopsy was performed in 181 patients, showing signs of small fibers neuropathy in 169 (93.4%). Positive, yet preliminary, results were observed in patients receiving periodic vitamin supplementation. Conclusions Over one third of PD patients in treatment with l-dopa may develop PNP, with a significantly higher prevalence of acute and subacute forms in those receiving LCIG. Pathogenic mechanisms remain unclear, but possibly related to a complex interplay between peripheral neurodegenerative processes and l-dopa neurotoxic metabolites. Prospective, randomized, clinical trials are required to identify factors associated with the onset and progression of PD-associated PNP and clarify the protective role of B-group vitamin supplementation.
Collapse
Affiliation(s)
- Alberto Romagnolo
- Department of Neuroscience "Rita Levi Montalcini" University of Turin Torino Italy
| | - Aristide Merola
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology University of Cincinnati Cincinnati Ohio USA
| | - Carlo Alberto Artusi
- Department of Neuroscience "Rita Levi Montalcini" University of Turin Torino Italy
| | | | - Maurizio Zibetti
- Department of Neuroscience "Rita Levi Montalcini" University of Turin Torino Italy
| | - Leonardo Lopiano
- Department of Neuroscience "Rita Levi Montalcini" University of Turin Torino Italy
| |
Collapse
|
57
|
Kaur H, Bhatia K, Yelam A, Shukla A, Asher I, Govindarajan R. Infusion-rate-dependent Acute Neuropathic Pain with Duopa in a Patient with Parkinson's Disease and Pre-existing Neuropathy. Cureus 2018; 10:e3055. [PMID: 30276034 PMCID: PMC6160297 DOI: 10.7759/cureus.3055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The association of peripheral neuropathy (PN) with the cumulative use of levodopa, presumably by vitamin B12 deficiency, has been well-documented in the literature. However, less is known about the L-dopa infusion-rate-dependent precipitation of pre-existing peripheral neuropathy. We report an unusual case of Parkinson’s disease in a patient who presented with acute exacerbations of pre-existing peripheral neuropathy on exceeding certain rates of continuous Carbidopa-Levodopa Infusions (CLI). We present a case of a 68-year-old gentleman with a 20-year history of idiopathic Parkinson's disease with bilateral subthalamic nucleus deep brain stimulation who was started on Duopa therapy for worsening dyskinesias. Workup before initiating Duopa was significant for idiopathic sensorimotor axonal polyneuropathy, although the symptoms were well controlled with medications. Subsequently, the patient developed severe neuropathic pain within 24 hours of initiating Duopa characterized by burning and stinging in his feet. Symptoms resolved within four hours of reducing the continuous infusion rates without modifying the bolus doses. The patient was doing well with extra bolus doses to manage off periods with no further recurrence of symptoms. With the support of this case report, we would like to conclude that the acute worsening of neuropathic pain with infusion rate or duration of treatment might limit the clinical benefits of Duopa and adds to the expanding spectrum of neurotoxic side effects associated with this therapy. Further prospective studies are needed to monitor the acute worsening of neuropathic pain in patients with pre-existing PN, after initiating CLI and its association with doses of levodopa/carbidopa.
Collapse
Affiliation(s)
- Harleen Kaur
- Neurology, Univeristy of Missouri, Columbia, USA
| | - Kunal Bhatia
- Neurology, University of Missouri, Columbia, USA
| | | | | | - Irving Asher
- Neurology, University of Missouri, Columbia, USA
| | | |
Collapse
|
58
|
Bourdon AK, Spano GM, Marshall W, Bellesi M, Tononi G, Serra PA, Baghdoyan HA, Lydic R, Campagna SR, Cirelli C. Metabolomic analysis of mouse prefrontal cortex reveals upregulated analytes during wakefulness compared to sleep. Sci Rep 2018; 8:11225. [PMID: 30046159 PMCID: PMC6060152 DOI: 10.1038/s41598-018-29511-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
By identifying endogenous molecules in brain extracellular fluid metabolomics can provide insight into the regulatory mechanisms and functions of sleep. Here we studied how the cortical metabolome changes during sleep, sleep deprivation and spontaneous wakefulness. Mice were implanted with electrodes for chronic sleep/wake recording and with microdialysis probes targeting prefrontal and primary motor cortex. Metabolites were measured using ultra performance liquid chromatography-high resolution mass spectrometry. Sleep/wake changes in metabolites were evaluated using partial least squares discriminant analysis, linear mixed effects model analysis of variance, and machine-learning algorithms. More than 30 known metabolites were reliably detected in most samples. When used by a logistic regression classifier, the profile of these metabolites across sleep, spontaneous wake, and enforced wake was sufficient to assign mice to their correct experimental group (pair-wise) in 80-100% of cases. Eleven of these metabolites showed significantly higher levels in awake than in sleeping mice. Some changes extend previous findings (glutamate, homovanillic acid, lactate, pyruvate, tryptophan, uridine), while others are novel (D-gluconate, N-acetyl-beta-alanine, N-acetylglutamine, orotate, succinate/methylmalonate). The upregulation of the de novo pyrimidine pathway, gluconate shunt and aerobic glycolysis may reflect a wake-dependent need to promote the synthesis of many essential components, from nucleic acids to synaptic membranes.
Collapse
Affiliation(s)
- Allen K Bourdon
- Department of Chemistry, University of Tennessee, Knoxville, TN, United States
| | - Giovanna Maria Spano
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - William Marshall
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - Michele Bellesi
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - Pier Andrea Serra
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Helen A Baghdoyan
- Department of Anesthesiology and Psychology, University of Tennessee, Knoxville, TN, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Ralph Lydic
- Department of Anesthesiology and Psychology, University of Tennessee, Knoxville, TN, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN, United States. .,Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN, United States.
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States.
| |
Collapse
|
59
|
The influence of levodopa, entacapone and homocysteine on prevalence of polyneuropathy in patients with Parkinson's disease. J Neurol Sci 2018; 392:28-31. [PMID: 30097148 DOI: 10.1016/j.jns.2018.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
|
60
|
Salama M, Shalash A, Magdy A, Makar M, Roushdy T, Elbalkimy M, Elrassas H, Elkafrawy P, Mohamed W, Abou Donia MB. Tubulin and Tau: Possible targets for diagnosis of Parkinson's and Alzheimer's diseases. PLoS One 2018; 13:e0196436. [PMID: 29742117 PMCID: PMC5942772 DOI: 10.1371/journal.pone.0196436] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 04/12/2018] [Indexed: 11/29/2022] Open
Abstract
Neurodegenerative diseases including Alzheimer’s disease (AD) and Parkinson’s disease (PD) are characterized by progressive neuronal loss and pathological accumulation of some proteins. Developing new biomarkers for both diseases is highly important for the early diagnosis and possible development of neuro-protective strategies. Serum antibodies (AIAs) against neuronal proteins are potential biomarkers for AD and PD that may be formed in response to their release into systemic circulation after brain damage. In the present study, two AIAs (tubulin and tau) were measured in sera of patients of PD and AD, compared to healthy controls. Results showed that both antibodies were elevated in patients with PD and AD compared to match controls. Curiously, the profile of elevation of antibodies was different in both diseases. In PD cases, tubulin and tau AIAs levels were similar. On the other hand, AD patients showed more elevation of tau AIAs compared to tubulin. Our current results suggested that AIAs panel could be able to identify cases with neuro-degeneration when compared with healthy subjects. More interestingly, it is possible to differentiate between PD and AD cases through identifying specific AIAs profile for each neurodegenerative states.
Collapse
Affiliation(s)
- Mohamed Salama
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- * E-mail:
| | - Ali Shalash
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alshimaa Magdy
- Biochemistry Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Marianne Makar
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tamer Roushdy
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mahmoud Elbalkimy
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hanan Elrassas
- Okasha Institute of Psychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Wael Mohamed
- Department of Pharmacology, Faculty of Medicine, Menoufia University, Shebeen Elkoum, Egypt
- Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan Pahang, Malaysia
| | - Mohamed B. Abou Donia
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
61
|
Christine CW, Auinger P, Joslin A, Yelpaala Y, Green R. Vitamin B12 and Homocysteine Levels Predict Different Outcomes in Early Parkinson's Disease. Mov Disord 2018; 33:762-770. [DOI: 10.1002/mds.27301] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/12/2017] [Accepted: 12/31/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
| | - Peggy Auinger
- Center for Human Experimental Therapeutics; University of Rochester; Rochester New York USA
| | - Amelia Joslin
- Department of Pathology and Laboratory Medicine; UC Davis; Davis California USA
| | - Yuora Yelpaala
- Department of Pathology and Laboratory Medicine; UC Davis; Davis California USA
| | - Ralph Green
- Department of Pathology and Laboratory Medicine; UC Davis; Davis California USA
| | | |
Collapse
|
62
|
Coon EA, Cutsforth-Gregory JK, Benarroch EE. Neuropathology of autonomic dysfunction in synucleinopathies. Mov Disord 2018; 33:349-358. [DOI: 10.1002/mds.27186] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/30/2017] [Accepted: 09/10/2017] [Indexed: 12/16/2022] Open
|
63
|
Coon EA, Low PA. Thermoregulation in Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2018; 157:715-725. [DOI: 10.1016/b978-0-444-64074-1.00043-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
64
|
Rispoli V, Simioni V, Capone JG, Golfrè Andreasi N, Preda F, Sette E, Tugnoli V, Sensi M. Peripheral neuropathy in 30 duodopa patients with vitamins B supplementation. Acta Neurol Scand 2017; 136:660-667. [PMID: 28608472 DOI: 10.1111/ane.12783] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2017] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Peripheral neuropathy (PN) is a significant concern and potential cause of withdrawal in patients with Parkinson's disease (PD) treated with Levodopa/Carbidopa Intestinal Gel (LCIG) infusion. Vitamin B deficiency and/or hyperhomocysteinemia levodopa-related are considered possible causative factors. In this study, we evaluated PN incidence in LCIG-PD patients treated since the beginning of infusion with vitamins B supplementation. MATERIALS & METHODS In this prospective open-label pilot study, 30 consecutive patients with PD on LCIG infusion were evaluated with clinical, neurophysiological, and biochemical assessments for a mean follow-up of 42.4 months (range 24-72). All evaluations were repeated every 6 months. RESULTS At baseline, 21 of 30 presented no signs or symptoms of PN, and 9 of 30 had pre-existing chronic PN. In whole population, a progressive worsening in nerve conduction studies of sural sensory and peroneal motor nerves was observed during the long-term follow-up. 4 of 21 patients, with normal clinical, electrophysiological assessment at baseline, developed distal symmetrical axonal polyneuropathy that remained asymptomatic during the long-term follow-up. Patients with pre-existing PN (9 of 30) showed a mild worsening of electrophysiological features during the period of observation. In none PN was cause of discontinuation of LCIG therapy. No incident cases of acute-subacute PN were documented. No correlation was found with age, sex, Levodopa dosage, duration of levodopa exposure, and homocysteine plasma levels. CONCLUSION In this consecutive series of 30 patients with PD on LCIG infusion, with early and continuous vitamins B integration, we observed a low rate (19%) of new onset peripheral polyneuropathy that remained stable after long-term follow-up. Larger studies, controlled, with blinded evaluation, are needed to confirm these findings.
Collapse
Affiliation(s)
- V. Rispoli
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| | - V. Simioni
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| | - J. G. Capone
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| | - N. Golfrè Andreasi
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| | - F. Preda
- Neurology Department; Ospedale Bufalini; Cesena Italy
| | - E. Sette
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| | - V. Tugnoli
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| | - M. Sensi
- Neurology Department; Az. Ospedaliero-Universitaria Arcispedale Sant'Anna; Cona Ferrara Italy
| |
Collapse
|
65
|
Andréasson M, Brodin L, Laffita-Mesa JM, Svenningsson P. Correlations Between Methionine Cycle Metabolism, COMT Genotype, and Polyneuropathy in L-Dopa Treated Parkinson’s Disease: A Preliminary Cross-Sectional Study. JOURNAL OF PARKINSONS DISEASE 2017; 7:619-628. [DOI: 10.3233/jpd-171127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mattias Andréasson
- Department of Neurology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lovisa Brodin
- Department of Neurology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - José Miguel Laffita-Mesa
- Department of Clinical Neuroscience, Translational Neuropharmacology, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Per Svenningsson
- Department of Neurology, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Clinical Neuroscience, Translational Neuropharmacology, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
66
|
Nolano M, Provitera V, Manganelli F, Iodice R, Stancanelli A, Caporaso G, Saltalamacchia A, Califano F, Lanzillo B, Picillo M, Barone P, Santoro L. Loss of cutaneous large and small fibers in naive and l-dopa-treated PD patients. Neurology 2017; 89:776-784. [PMID: 28747449 DOI: 10.1212/wnl.0000000000004274] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To study small and large fiber pathology in drug-naive and l-dopa-treated patients affected by Parkinson disease (PD) in early phases, before the occurrence of neuropathic electrophysiologic abnormalities. METHODS We enrolled 85 patients with idiopathic PD (male/female 49/36, age 61.3 ± 9.7 years) without electrophysiologic signs of neuropathy, including 48 participants naive to l-dopa treatment. All patients underwent clinical, functional, and morphologic assessment of sensory and autonomic nerves through dedicated questionnaires, quantitative sensory testing, sympathetic skin response, dynamic sweat test, and punch biopsies from glabrous and hairy skin. Sensory and autonomic innervation was visualized with specific antibodies and analyzed by confocal microscopy. Data were compared with those obtained from sex- and age-comparable healthy controls. In 35 patients, skin biopsies were performed bilaterally to evaluate side-to-side differences. RESULTS Intraepidermal nerve fiber density was lower in patients compared to controls in all the examined sites (p < 0.001). The loss was higher in the more affected side (p < 0.01). A loss of autonomic nerves to vessels, sweat glands, and arrector pili muscles and of Meissner corpuscles and their myelinated endings in glabrous skin was found (p < 0.001). Patients showed increased tactile and thermal thresholds, impairment of mechanical pain perception, and reduced sweat output (p < 0.001). The naive and l-dopa-treated groups differed only for Meissner corpuscle density (p < 0.001). CONCLUSIONS Both large and small fiber pathology occurs in the early stages of PD and may account for the sensory and autonomic impairment. l-Dopa affects the 2 populations of fibers differently.
Collapse
Affiliation(s)
- Maria Nolano
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy.
| | - Vincenzo Provitera
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Fiore Manganelli
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Rosa Iodice
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Annamaria Stancanelli
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Giuseppe Caporaso
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Annamaria Saltalamacchia
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Francesca Califano
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Bernardo Lanzillo
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Marina Picillo
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Paolo Barone
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Lucio Santoro
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| |
Collapse
|
67
|
Park JS, Park D, Ko PW, Kang K, Lee HW. Serum methylmalonic acid correlates with neuropathic pain in idiopathic Parkinson's disease. Neurol Sci 2017; 38:1799-1804. [PMID: 28726051 DOI: 10.1007/s10072-017-3056-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/01/2017] [Indexed: 10/19/2022]
Abstract
Recent studies have shown a relatively higher prevalence of peripheral neuropathy in idiopathic Parkinson's disease (IPD). The hypothesis is that prolonged levodopa exposure causes vitamin B12 deficiency, which leads to peripheral neuropathy. The aim of our study was to find the relationship between vitamin B12 and its precursor methylmalonic acid (MMA) in IPD patients with neuropathic pain. We performed a cross-sectional study by enrolling consecutive 43 patients who were clinically tested positive for F-18 FP-CIT PET and 15 patients were diagnosed with peripheral neuropathy according to the Toronto clinical scoring system (TCSS). The severity of neuropathic pain was evaluated using total neuropathy scale, revised (TNSr), and Korean Neuropathic Pain Questionnaire (KNPQ). The correlations between age, IPD duration, levodopa equivalent dose (LED), UPDRS III, vitamin B12, MMA, and homocysteine levels were assessed. The prevalence rate of peripheral neuropathy in IPD patients was 35%. Among the serums assessed, MMA levels showed a positive correlation to TNSr and KNPQ in the IPD patients with peripheral neuropathy (TNSr r = 0.882, p < 0.001, KNPQ r = 0.710, p = 0.004), while Vitamin B12 and homocysteine showed no statistically significant correlation. Our study showed a prevalence of peripheral neuropathy in 35% of Korean IPD patients. The serum MMA positively correlated with the severity of neuropathic pain and this can be used as a useful marker in assessment of peripheral neuropathy in Parkinson's disease.
Collapse
Affiliation(s)
- Jin-Sung Park
- Department of Neurology, Kyungpook National University, School of Medicine, Daegu, South Korea
| | - Donghwi Park
- Department of Rehabilitation Medicine, Daegu Fatima Hospital, Daegu, South Korea
| | - Pan-Woo Ko
- Department of Neurology, Kyungpook National University, School of Medicine, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Kyunghun Kang
- Department of Neurology, Kyungpook National University, School of Medicine, Daegu, South Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Ho-Won Lee
- Department of Neurology, Kyungpook National University, School of Medicine, Daegu, South Korea. .,Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
68
|
Zis P, Grünewald RA, Chaudhuri RK, Hadjivassiliou M. Peripheral neuropathy in idiopathic Parkinson's disease: A systematic review. J Neurol Sci 2017; 378:204-209. [PMID: 28566165 DOI: 10.1016/j.jns.2017.05.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Parkinson's disease (PD) has been associated with peripheral neuropathy (PN). PN has been demonstrated in some rare genetic forms of PD (e.g. PARK2 mutations) but has also been linked to levodopa exposure. OBJECTIVE The aim of this systematic review is to clarify any evidence of peripheral nervous system involvement in idiopathic PD. METHODS A systematic computer-based literature search was conducted on PubMed database. FINDINGS The pooled estimate of the prevalence of large fiber PN in PD was 16.3% (based on 1376 patients). The pooled estimate of the prevalence of biopsy-proven small fiber neuropathy was 56.9% (based on 72 patients). Large fiber PN in PD is in the majority of cases distal, symmetrical, axonal and predominantly sensory. There are, however, few reports of chronic idiopathic demyelinating polyneuropathy and very occasional cases of acute neuropathies. Although nerve conduction studies have been performed in the majority of the studies, they included only a limited number of nerves, mainly in the lower limbs. There is little evidence to support a direct link between levodopa treatment and the development of PN in idiopathic PD. In the majority of the cases PN has been linked to abnormalities in vitamin B12, methylmalonic acid or fasting homocysteine levels. Additional aetiological risk factors for PN may be responsible for any apparent link between PD and PN. CONCLUSIONS Large-scale prospective studies with long-term follow-up with detailed baseline assessments are needed in order to understand the natural history of PN in PD, both on clinical and neurophysiological parameters.
Collapse
Affiliation(s)
- Panagiotis Zis
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; University of Sheffield, UK.
| | | | - Ray Kallol Chaudhuri
- National Parkinson Foundation International Centre of Excellence, King's College London and Kings College Hospital NHS Foundation Trust, London, UK
| | - Marios Hadjivassiliou
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; University of Sheffield, UK
| |
Collapse
|
69
|
Merola A, Rosso M, Romagnolo A, Comi C, Fasano A, Zibetti M, Lopez-Castellanos JR, Cocito D, Lopiano L, Espay AJ. Peripheral neuropathy as marker of severe Parkinson's disease phenotype. Mov Disord 2017; 32:1256-1258. [PMID: 28582598 DOI: 10.1002/mds.27025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/01/2017] [Accepted: 04/03/2017] [Indexed: 11/10/2022] Open
Affiliation(s)
- Aristide Merola
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michela Rosso
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Alberto Romagnolo
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, Section of Neurology, University of Piemonte Orientale, Novara, Italy
| | - Alfonso Fasano
- Division of Neurology, Movement Disorders Center, University of Toronto Canada Morton and Gloria Shulman Movement Disorders Centre and the Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, UHN, University of Toronto, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto, Ontario, Canada
| | - Maurizio Zibetti
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - José Ricardo Lopez-Castellanos
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Dario Cocito
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Leonardo Lopiano
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Alberto J Espay
- Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
70
|
Loens S, Chorbadzhieva E, Kleimann A, Dressler D, Schrader C. Effects of levodopa/carbidopa intestinal gel versus oral levodopa/carbidopa on B vitamin levels and neuropathy. Brain Behav 2017; 7:e00698. [PMID: 28523235 PMCID: PMC5434198 DOI: 10.1002/brb3.698] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/15/2017] [Accepted: 03/03/2017] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES To determine the possible interactions between levodopa therapy and plasma levels of B vitamins in patients with advanced idiopathic Parkinson's disease (IPD) in the context of either oral levodopa therapy or levodopa/carbidopa intestinal gel (LCIG). Secondly, to determine the prevalence of neuropathy and its relation to plasma levels of B vitamins and homocysteine. METHODS Medication doses, neurographies, and serum levels of pyridoxine, cobalamin, folate, and homocysteine of eight LCIG and 13 orally treated advanced IPD patients matched for age, Hoehn & Yahr stage, and UPRDS III were collected. This data was analyzed for correlation with daily levodopa dose (LDD). RESULTS LICG patients had a longer disease duration and higher LDD. All LCIG patients and most orally treated patients had sensorimotor axonal polyneuropathy. Of all plasma vitamin levels, pyridoxine was decreased most and significantly lower in the LCIG group. Cobalamin and folate, however, were within the lower reference range, and homocysteine highly elevated, all without any significant difference between both groups. LDD correlated significantly with pyridoxine deficiency (p = .02) irrespective of the route of application and with hyperhomocysteinemia in the LCIG group (p = .03). At LDDs above 2,000 mg, pyridoxine deficiency was almost always detectable. CONCLUSIONS Pyridoxine deficiency and hyperhomocysteinemia are dependent on the daily levodopa/carbidopa dose, while levels of cobalamin and folate are not. The mode of application of levodopa/carbidopa has no impact on B-vitamin levels. Neuropathy is very frequent in advanced IPD; however, it remains to be investigated further whether neuropathy is more frequent in LCIG than in orally levodopa/carbidopa-treated advanced IPD patients.
Collapse
Affiliation(s)
- Sebastian Loens
- Department of Neurology and Clinical Neurophysiology Hannover Medical School Hannover Germany
| | - Elena Chorbadzhieva
- Department of Neurology and Clinical Neurophysiology Hannover Medical School Hannover Germany
| | - Alexandra Kleimann
- Department of Psychiatry Social Psychiatry and Psychotherapy Hannover Medical School Hannover Germany
| | - Dirk Dressler
- Department of Neurology and Clinical Neurophysiology Hannover Medical School Hannover Germany
| | - Christoph Schrader
- Department of Neurology and Clinical Neurophysiology Hannover Medical School Hannover Germany
| |
Collapse
|
71
|
Levodopa/carbidopa intestinal gel (LCIG) infusion as mono- or combination therapy. J Neural Transm (Vienna) 2017; 124:1005-1013. [PMID: 28229223 DOI: 10.1007/s00702-017-1698-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/14/2017] [Indexed: 01/16/2023]
Abstract
Levodopa/carbidopa intestinal gel (LCIG) infusion is an effective escalating therapy in patients with Parkinson disease (PD) suffering from motor fluctuations and dyskinesia. Levodopa/carbidopa given continuously as infusion provides an optimized application of the most effective and best tolerable antiparkinsonian drug. It has been proven to have a superior motor effect compared with oral levodopa and to improve also non-motor symptoms. However, invasiveness, discomfort resulting from carrying an external device, and side effects associated with the way of administration limit its application in PD patients. At present, there are no guidelines that delineate to which patients LCIG should be offered as monotherapy, in combination with oral and/or transdermal medication, or as additional therapy to deep brain stimulation (DBS). Based on clinical studies, we propose an expert consensus for neurologists addressing the question when LCIG therapy should be recommended and in which cases LCIG infusion is suggested in combination with other antiparkinsonian drugs and/or DBS. We describe how LCIG should be initiated and what we consider necessary for clinical follow-up. We suggest an algorithm facilitating decision-making with respect to the currently available invasive PD therapies, namely infusion with subcutaneous apomorphine, LCIG, and DBS.
Collapse
|
72
|
Mylius V, Teepker M, Zouari HG, Beer S, Schepelmann K, Möller JC. Clinical correlates of sural neurography impairment within normal limits in patients with Parkinsons disease. Neurophysiol Clin 2017; 47:83-84. [PMID: 28162844 DOI: 10.1016/j.neucli.2017.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/05/2017] [Indexed: 11/18/2022] Open
Affiliation(s)
- Veit Mylius
- Department of Neurology, Philipps University, Marburg, Germany; Center for Neurorehabilitation, Valens, Switzerland.
| | - Michael Teepker
- Department of Neurology, Philipps University, Marburg, Germany
| | - Hela G Zouari
- Service d'explorations fonctionnelles, CHU Habib Bourguiba, Sfax, Tunisia
| | - Serafin Beer
- Center for Neurorehabilitation, Valens, Switzerland
| | | | - Jens C Möller
- Department of Neurology, Philipps University, Marburg, Germany; Parkinson center, Center for Neurological Rehabilitation, Zihlschlacht, Switzerland
| |
Collapse
|
73
|
Newer anti-epileptic drugs, vitamin status and neuropathy: A cross-sectional analysis. Rev Neurol (Paris) 2017; 173:62-66. [DOI: 10.1016/j.neurol.2016.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/29/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
|
74
|
Senek M, Nielsen EI, Nyholm D. Levodopa-entacapone-carbidopa intestinal gel in Parkinson's disease: A randomized crossover study. Mov Disord 2016; 32:283-286. [PMID: 27987231 DOI: 10.1002/mds.26855] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/23/2016] [Accepted: 10/02/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The addition of oral entacapone to levodopa-carbidopa intestinal gel treatment leads to less conversion of levodopa to 3-O-methyldopa, thereby increasing levodopa plasma concentration. The objective of this study was to compare systemic levodopa exposure of the newly developed levodopa-entacapone-carbidopa intestinal gel after a 20% dose reduction with levodopa exposure after the usual levodopa-carbidopa intestinal gel dose in a randomized crossover trial in advanced Parkinson's disease patients. METHODS In this 48-hour study, 11 patients treated with levodopa-carbidopa intestinal gel were randomized to a treatment sequence. Blood samples were drawn at prespecified times, and patient motor function was assessed according to the treatment response scale. RESULTS Systemic exposure of levodopa did not differ significantly between treatments (ratio, 1.10 [95% confidence interval, 0.951-1.17]). Treatment response scale scores did not significantly differ between treatments (P = 0.84). CONCLUSIONS Levodopa-entacapone-carbidopa intestinal gel allowed a lower amount of levodopa administration and was well tolerated. Long-term studies are needed to confirm the results. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marina Senek
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Elisabet I Nielsen
- Department of Pharmaceutical Biosciences, Pharmacometrics, Uppsala University, Uppsala, Sweden
| | - Dag Nyholm
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
75
|
Virhammar J, Nyholm D. Levodopa-carbidopa enteral suspension in advanced Parkinson's disease: clinical evidence and experience. Ther Adv Neurol Disord 2016; 10:171-187. [PMID: 28344656 DOI: 10.1177/1756285616681280] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The duration of action of oral levodopa becomes shorter as Parkinson's disease (PD) progresses. Patients with advanced PD may develop potentially disabling motor fluctuations and abnormal involuntary movement (dyskinesia), which cannot be managed with optimized oral or transdermal PD medications. The progressively worsening symptoms can have a substantial impact on the patient quality of life (QoL). Levodopa-carbidopa intestinal gel (LCIG) is delivered continuously via a percutaneous endoscopic gastrostomy with a jejunal extension (PEG-J). LCIG is licensed for the treatment of levodopa-responsive advanced PD in individuals experiencing severe motor fluctuations and dyskinesia when available combinations of antiparkinsonian medications have not given satisfactory results. Initial evidence for the efficacy and tolerability of LCIG came from a number of small-scale studies, but recently, three prospective studies have provided higher quality evidence. A 12-week double-blind comparison of LCIG with standard levodopa therapy, a 52-week open-label study extension of the double-blind study, and a 54-week open-label safety study, demonstrated significant improvements in 'off' time and 'on' time without troublesome dyskinesia, and QoL measures that were maintained in the longer term. There are also observations that LCIG may be effective treatment for nonmotor symptoms (NMS) although the evidence is limited. There is a need for further research on the efficacy of LCIG in reducing NMS, dyskinesia and improving QoL. This review surveys the clinical evidence for the effectiveness and tolerability of LCIG in the management of advanced PD and highlights some practical considerations to help optimize treatment.
Collapse
Affiliation(s)
- Johan Virhammar
- Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Dag Nyholm
- Department of Neuroscience, Neurology, Uppsala University Hospital, 751 85 Uppsala, Sweden
| |
Collapse
|
76
|
Anti-MAG autoantibodies are increased in Parkinson's disease but not in atypical parkinsonism. J Neural Transm (Vienna) 2016; 124:209-216. [PMID: 27766424 DOI: 10.1007/s00702-016-1632-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 10/09/2016] [Indexed: 01/16/2023]
Abstract
There is emerging evidence that glial cells are involved in the neuropathological process in Parkinson's disease (PD) in addition to degeneration of neuronal structures. Recently, we confirmed the presence of an adaptive immune response against different glial-derived antigens in PD, with a possible role of anti-MAG, anti-MBP and anti-PLP antibodies in the disease progression. The aim of the present study was to assess humoral response against myelin-associated glycoprotein (MAG) in patients with parkinsonism (both idiopathic and atypical) to check whether these antibodies could serve as biomarkers of PD, its severity and progression. Anti-MAG autoantibodies were measured by an ELISA system in 99 PD patients, 33 atypical parkinsonism patients, and 36 control subjects. In PD patients, anti-MAG IgM autoantibodies were significantly higher in comparison to healthy control subjects (p = 0.038). IgM anti-MAG autoantibodies titers were also significantly higher in the whole group of patients with parkinsonism (either idiopathic or atypical) in comparison to healthy control subjects (1.88 ± 0.84 vs 1.70 ± 1.19, p = 0.017). This difference was mainly driven by the PD group, as the atypical parkinsonism group did not differ significantly from the control group in anti-MAG antibody levels (p = 0.51). A negative correlation between anti-MAG levels and disease duration was found in PD patients. Our study provides evidence for an increased production of autoantibodies against a protein of glial origin in PD. The negative correlation between anti-MAG antibodies and disease duration may suggest possible involvement of the immune system in disease progression. Increasing evidence that glia are involved in the neurodegenerative process to a greater extent than previously thought may turn out be useful in the search for biomarkers of the neurodegenerative process in PD.
Collapse
|
77
|
Schwab AJ, Ebert AD. Neurite Aggregation and Calcium Dysfunction in iPSC-Derived Sensory Neurons with Parkinson's Disease-Related LRRK2 G2019S Mutation. Stem Cell Reports 2016; 5:1039-1052. [PMID: 26651604 PMCID: PMC4682343 DOI: 10.1016/j.stemcr.2015.11.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 11/11/2015] [Accepted: 11/12/2015] [Indexed: 01/15/2023] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most-common genetic determinants of Parkinson’s disease (PD). The G2019S mutation is detected most frequently and is associated with increased kinase activity. Whereas G2019S mutant dopamine neurons exhibit neurite elongation deficits, the effect of G2019S on other neuronal subtypes is unknown. As PD patients also suffer from non-motor symptoms that may be unrelated to dopamine neuron loss, we used induced pluripotent stem cells (iPSCs) to assess morphological and functional properties of peripheral sensory neurons. LRRK2 G2019S iPSC-derived sensory neurons exhibited normal neurite length but had large microtubule-containing neurite aggregations. Additionally, LRRK2 G2019S iPSC-derived sensory neurons displayed altered calcium dynamics. Treatment with LRRK2 kinase inhibitors resulted in significant, but not complete, morphological and functional rescue. These data indicate a role for LRRK2 kinase activity in sensory neuron structure and function, which when disrupted, may lead to sensory neuron deficits in PD. LRRK2 iPSC sensory neurons show neurite aggregations and abnormal calcium dynamics LRRK2 iPSC sensory neuron defects are distinct from the dopamine neuron defects Kinase inhibition of LRRK2 partially restored sensory neuron structure and function Abnormal sensory neuron phenotypes may relate to non-motor symptoms observed in PD
Collapse
Affiliation(s)
- Andrew J Schwab
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
78
|
Polyneuropathy in levodopa-treated Parkinson's patients. J Neurol Sci 2016; 371:36-41. [PMID: 27871444 DOI: 10.1016/j.jns.2016.09.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/13/2016] [Accepted: 09/30/2016] [Indexed: 11/22/2022]
Abstract
Recently published studies show that the prevalence of polyneuropathy (PNP) is higher in patients with Parkinson's disease (PD) than in age-matched controls. Its pathogenesis, however is a matter of controversy. The major hypothesis is the toxicity of high concentrations of homocysteine (Hcy) possibly related to levodopa (LD) therapy. The aim of the present study was to determine the prevalence of PNP, independent of other etiologies, and to determine the relationship to demographic and clinical factors in LD-treated Parkinson's patients. A total of 102 patients (51 patients with PD and 51 sex- and age-matched healthy controls) were enrolled in the study. The presence of any risk factors for PNP, ascertained from the history and laboratory tests, was an exclusion criterion. The Toronto Clinical Scoring System (TCSS) was used for clinical assessment of PNP. The objective assessment was based on electroneurography (ENG) studies in which motor nerves (peroneal and tibial nerves) as well as sensory nerves (sural and superficial peroneal nerves) were bilaterally examined. The severity of the disease was determined using the UPDRS scale (Unified Parkinson's Disease Rating Scale) and the Hoehn-Yahr (H-Y) scale. In the PD group, the clinical and neurophysiological indicators of PNP, manifested as a symmetrical and predominantly sensory axonal neuropathy, were more frequent then in the control group and observed in 43.1% vs. 13.7% and 15.7% vs. 2% of subjects respectively. The presence of PNP correlated with age and the severity of PD. Patients with PD and PNP had a higher level of Hcy as compared to PD patients without PNP, however the difference was not statistically significant. The frequency of PNP in PD patients is higher than in controls. The characteristics and discrepancy between the number of patients with clinical and ENG detected PNP may suggest the small fiber neuropathy (SFN) as the dominant form of neuropathy in PD patients.
Collapse
|
79
|
Electrophysiological assessment of nociception in patients with Parkinson's disease: A multi-methods approach. J Neurol Sci 2016; 368:59-69. [DOI: 10.1016/j.jns.2016.06.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 11/21/2022]
|
80
|
de Araújo DF, de Melo Neto AP, Oliveira ÍSC, Brito BS, de Araújo IT, Barros IS, Lima JWO, Horta WG, Gondim FDAA. Small (autonomic) and large fiber neuropathy in Parkinson disease and parkinsonism. BMC Neurol 2016; 16:139. [PMID: 27530902 PMCID: PMC4988006 DOI: 10.1186/s12883-016-0667-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 08/10/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent studies have reported that peripheral neuropathy (PN) is common in patients with Parkinson's disease (PD) and raised the possibility that levodopa neurotoxicity is the main culprit. METHODS We evaluated the presence of large & small (autonomic) fiber PN in 54 consecutive patients with PD or parkinsonism in a tertiary outpatient clinic from Brazil. Initial PN screening consisted of history/neurological exam and skin wrinkling test (SWT). In addition, we also performed Nerve conduction studies/Electromyography (NCS/EMG) in all patients with PN signs/symptoms and/or abnormal SWT. RESULTS Thirty eight patients with PD (10 women, mean age: 63 ± 2.1 years, P < 0.05 versus parkinsonism, mean disease duration: 8 ± 0.8 years) and 16 patients with other forms of parkinsonism [7 women, mean age: 50.1 ± 3.9 years, mean disease duration: 6.9 ± 1.1 years] completed clinical neuromuscular evaluation. SWT was performed in 48 patients (33 PD, 15 parkinsonism). In the PD group, SWT was abnormal in 57.6% of the tested patients (comprising 50% of all PD patients). In the parkinsonism group, SWT was abnormal in 37.5% (comprising 35.3% of all parkinsonism patients). NCS/EMG was performed in 39 patients (26 PD and 13 parkinsonism). Twelve out of the 26 PD (34.2% of all PD) and 4 out of the 13 parkinsonism (23.5% of all parkinsonism) had abnormal NCS/EMG results. Neuropathy prevalence was similar in PD and parkinsonism groups as detected either by NCS/EMG or SWT. CONCLUSIONS Large fiber and small (autonomic) fiber PN are common in patients with PD and parkinsonism. The etiology for the neuropathy was likely to be multifactorial and may be secondary to PD itself.
Collapse
Affiliation(s)
- Davi Farias de Araújo
- Federal University of Ceará (UFC), Professor Costa Mendes Street, 949, Fortaleza, Ceará, Brazil
| | | | | | - Beatriz Soares Brito
- Federal University of Ceará (UFC), Professor Costa Mendes Street, 949, Fortaleza, Ceará, Brazil
| | | | - Ingrid Sousa Barros
- Federal University of Ceará (UFC), Professor Costa Mendes Street, 949, Fortaleza, Ceará, Brazil
| | | | - Wagner Goes Horta
- Department of Internal Medicine, Neurology Division, Federal University of Ceará, Professor Costa Mendes Street, 1608, Fortaleza, Ceará, Brazil
| | - Francisco de Assis Aquino Gondim
- Department of Internal Medicine, Neurology Division, Federal University of Ceará, Professor Costa Mendes Street, 1608, Fortaleza, Ceará, Brazil.
| |
Collapse
|
81
|
De Rosa A, Tessitore A, Bilo L, Peluso S, De Michele G. Infusion treatments and deep brain stimulation in Parkinson's Disease: The role of nursing. Geriatr Nurs 2016; 37:434-439. [PMID: 27444659 DOI: 10.1016/j.gerinurse.2016.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/09/2016] [Accepted: 06/13/2016] [Indexed: 12/27/2022]
Abstract
Parkinson's Disease (PD) represents one of the most common neurodegenerative disorders in the elderly. PD is caused by a loss of dopaminergic cells in the substantia nigra pars compacta. The motor cardinal signs include a resting tremor, bradykinesia, rigidity and postural reflex impairment. Although levodopa represents the gold standard also in the advanced stage of the disease, over the years most patients develop disabling motor fluctuations, dyskinesias, and non-motor complications, which are difficult to manage. At this stage, more complex treatment approaches, such as infusion therapies (subcutaneous apomorphine and intraduodenal levodopa) and deep brain stimulation of the subthalamic nucleus or the globus pallidus internus should be considered. All three procedures require careful selection and good compliance of candidate patients. In particular, infusional therapies need adequate training both of caregivers and nursing staff in order to assist clinicians in the management of patients in the complicated stages of disease.
Collapse
Affiliation(s)
- Anna De Rosa
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy.
| | - Alessandro Tessitore
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Leonilda Bilo
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
| | - Silvio Peluso
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
| | - Giuseppe De Michele
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
82
|
McKeown MD, Peters RM, Pasman EP, McKeown MJ, Carpenter MG, Inglis JT. Plantar cutaneous function in Parkinson's disease patients ON and OFF L-dopa. Neurosci Lett 2016; 629:251-255. [PMID: 27424795 DOI: 10.1016/j.neulet.2016.07.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 10/21/2022]
Abstract
While Parkinson's disease (PD) is traditionally viewed as a motor disorder, there is mounting evidence that somatosensory function becomes affected as well. However, conflicting reports exist regarding whether plantar sensitivity is reduced in early-onset PD patients. Plantar sensitivity was assessed using monofilaments and a gold-standard, two-interval two-alternative forced choice vibrotactile detection task at both 30 and 250Hz. Lower-limb cutaneous reflexes were assessed by delivering continuous, sinusoidal vibration at 30 and 250Hz while recording muscle activity in Tibialis Anterior. We found no evidence of elevated plantar thresholds or dysfunctional lower-limb cutaneous reflexes in PD patients ON medication. We also found no acute effect of ceasing L-dopa intake on either plantar sensitivity or cutaneous reflexes. Our finding of intact cutaneous function in PD supports the further exploration of therapeutics that enhance plantar sensitivity to minimize postural instability, a source of considerable morbidity in this clinical population.
Collapse
Affiliation(s)
- Monica D McKeown
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Ryan M Peters
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada.
| | - Elizabeth P Pasman
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | - Martin J McKeown
- Department of Medicine, Neurology, University of British Columbia, Vancouver, BC, Canada; Pacific Parkinson's Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Mark G Carpenter
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada
| | - J Timothy Inglis
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
83
|
Cossu G, Ceravolo R, Zibetti M, Arca R, Ricchi V, Paribello A, Murgia D, Merola A, Romagnolo A, Nicoletti V, Palermo G, Mereu A, Lopiano L, Melis M, Abbruzzese G, Bonuccelli U. Levodopa and neuropathy risk in patients with Parkinson disease: Effect of COMT inhibition. Parkinsonism Relat Disord 2016; 27:81-4. [DOI: 10.1016/j.parkreldis.2016.04.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/17/2016] [Accepted: 04/18/2016] [Indexed: 12/19/2022]
|
84
|
Kesayan T, Lamb DG, Falchook AD, Williamson JB, Salazar L, Malaty IA, McFarland NR, Okun MS, Wagle Shukla A, Heilman KM. Abnormal tactile pressure perception in Parkinson's disease. J Clin Exp Neuropsychol 2016; 37:808-15. [PMID: 26313511 DOI: 10.1080/13803395.2015.1060951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND/OBJECTIVE Some of the behavioral disorders associated with Parkinson's disease (PD), such as the reduced magnitude of actions (hypometria) may be related to an impairment in cognitive disengagement. A reduced ability to disengage attention from previous sensory stimuli will alter perception with a reduced range of estimated stimulus magnitudes (contraction to the mean). To test this disengagement hypothesis, participants with PD were tested to learn whether they had abnormal sensory perception with overestimation of the relative magnitude of weaker tactile stimuli and underestimation of the relative magnitude of stronger tactile stimuli in relation to a reference stimulus. DESIGN/METHOD The participants were 12 people with PD and 12 healthy adults. Test stimuli were applied to the palm using Semmes-Weinstein monofilaments (SWM) of 6 magnitudes, 3 greater and 3 less than a standard stimulus. In each trial, after being stimulated with the reference (standard) stimulus, a test monofilament was applied, and the participant was asked to provide a numerical estimate of the magnitude of the second stimulus relative to the standard. RESULTS Compared to the control group, participants with PD overestimated the magnitudes of the tactile stimuli below the standard stimulus and underestimated the magnitudes of stimuli above the standard stimulus. CONCLUSIONS These results demonstrate that people with PD likely have a reduced ability to estimate the relative magnitudes of tactile sensory stimuli. Whereas deafferentation would alter perception in one direction, the impairment of these participants with PD may result from a disorder of disengagement, and disorders of disengagement are often due to frontal-executive dysfunction.
Collapse
Affiliation(s)
- Tigran Kesayan
- a Department of Neurology , University of Florida , Gainesville , FL , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Podgorny PJ, Suchowersky O, Romanchuk KG, Feasby TE. Evidence for small fiber neuropathy in early Parkinson's disease. Parkinsonism Relat Disord 2016; 28:94-9. [PMID: 27160569 DOI: 10.1016/j.parkreldis.2016.04.033] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is neurodegenerative movement disorder affecting primarily the central nervous system with several recognized non-motor symptoms that can occur at various stages of the disease. Recently it has been shown that patients with PD may be prone to peripheral nervous system pathology in the form of a peripheral neuropathy (PN). It is unclear if PN is an inherent feature of PD or if it is an iatrogenic effect of the mainstay PD treatment Levodopa. METHODS To determine if peripheral neuropathy occurs in early untreated PD we employed a case-control study design using gold standard tests for PN, including neurological examination according to the Utah Early Neuropathy Scale (UENS) and nerve conduction studies, as well as new, more sensitive and informative tests for PN including the skin biopsy and corneal confocal microscopy (CCM). RESULTS We studied 26 patients with PD and 22 controls using the neurological examination and nerve conduction studies (NCS) and found no significant difference between groups except for some reduced vibration sense in the PD group. Epidermal nerve densities in the skin biopsies were similar between our cohorts. However, using CCM - a more sensitive test and a surrogate marker of small fiber damage in PN, we found that patients with PD had significantly reduced corneal nerve fiber densities and lengths as compared to controls. CONCLUSIONS We conclude that our positive CCM results provide evidence of preclinical PN in newly diagnosed PD patients.
Collapse
Affiliation(s)
- Peter J Podgorny
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Oksana Suchowersky
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, Canada; Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| | - Kenneth G Romanchuk
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Thomas E Feasby
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
86
|
|
87
|
Santos-García D, Mir P, Cubo E, Vela L, Rodríguez-Oroz MC, Martí MJ, Arbelo JM, Infante J, Kulisevsky J, Martínez-Martín P. COPPADIS-2015 (COhort of Patients with PArkinson's DIsease in Spain, 2015), a global--clinical evaluations, serum biomarkers, genetic studies and neuroimaging--prospective, multicenter, non-interventional, long-term study on Parkinson's disease progression. BMC Neurol 2016; 16:26. [PMID: 26911448 PMCID: PMC4766717 DOI: 10.1186/s12883-016-0548-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/19/2016] [Indexed: 12/19/2022] Open
Abstract
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder causing motor and non-motor symptoms that can affect independence, social adjustment and the quality of life (QoL) of both patients and caregivers. Studies designed to find diagnostic and/or progression biomarkers of PD are needed. We describe here the study protocol of COPPADIS-2015 (COhort of Patients with PArkinson’s DIsease in Spain, 2015), an integral PD project based on four aspects/concepts: 1) PD as a global disease (motor and non-motor symptoms); 2) QoL and caregiver issues; 3) Biomarkers; 4) Disease progression. Methods/design Observational, descriptive, non-interventional, 5-year follow-up, national (Spain), multicenter (45 centers from 15 autonomous communities), evaluation study. Specific goals: (1) detailed study (clinical evaluations, serum biomarkers, genetic studies and neuroimaging) of a population of PD patients from different areas of Spain, (2) comparison with a control group and (3) follow-up for 5 years. COPPADIS-2015 has been specifically designed to assess 17 proposed objectives. Study population: approximately 800 non-dementia PD patients, 600 principal caregivers and 400 control subjects. Study evaluations: (1) baseline includes motor assessment (e.g., Unified Parkinson’s Disease Rating Scale part III), non-motor symptoms (e.g., Non-Motor Symptoms Scale), cognition (e.g., Parkinson’s Disease Cognitive Rating Scale), mood and neuropsychiatric symptoms (e.g., Neuropsychiatric Inventory), disability, QoL (e.g., 39-item Parkinson’s disease Quality of Life Questionnaire Summary-Index) and caregiver status (e.g., Zarit Caregiver Burden Inventory); (2) follow-up includes annual (patients) or biannual (caregivers and controls) evaluations. Serum biomarkers (S-100b protein, TNF-α, IL-1, IL-2, IL-6, vitamin B12, methylmalonic acid, homocysteine, uric acid, C-reactive protein, ferritin, iron) and brain MRI (volumetry, tractography and MTAi [Medial Temporal Atrophy Index]), at baseline and at the end of follow-up, and genetic studies (DNA and RNA) at baseline will be performed in a subgroup of subjects (300 PD patients and 100 control subjects). Study periods: (1) recruitment period, from November, 2015 to February, 2017 (basal assessment); (2) follow-up period, 5 years; (3) closing date of clinical follow-up, May, 2022. Funding: Public/Private. Discussion COPPADIS-2015 is a challenging initiative. This project will provide important information on the natural history of PD and the value of various biomarkers.
Collapse
Affiliation(s)
- Diego Santos-García
- Sección de Neurología, Complejo Hospitalario Universitario de Ferrol (CHUF), Hospital Arquitecto Marcide, c/Avenida La Residencia, s/n, 15405, Ferrol, Spain.
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, CSIC y Universidad de Sevilla, Sevilla, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sevilla, Spain.
| | - Esther Cubo
- Servicio de Neurología, Hospital Universitario de Burgos, Burgos, Spain.
| | - Lydia Vela
- Unidad de Neurología, Fundación Hospital de Alcorcón, Madrid, Spain.
| | | | - Maria José Martí
- Unidad de Parkinson y Trastornos del Movimiento, Servicio de Neurología, Instituto Clínico de Neurociencias, Hospital Clínic, Barcelona, Spain.
| | - José Matías Arbelo
- Unidad de Trastornos del Movimiento y enfermedad de Parkinson, Servicio de Neurología, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Jon Infante
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital Universitario Marqués de Valdecilla, Santander, Spain.
| | - Jaime Kulisevsky
- Unidad de Trastornos del Movimiento, Servicio de Neurología, Hospital de Sant Pau, Barcelona, Spain.
| | - Pablo Martínez-Martín
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Sevilla, Spain. .,Centro Nacional de Epidemiología, Instituto de Salud Carlos III, Madrid, Spain.
| | | |
Collapse
|
88
|
Cossu G, Melis M. The peripheral nerve involvement in Parkinson Disease: A multifaceted phenomenon. Parkinsonism Relat Disord 2016; 25:17-20. [PMID: 26857398 DOI: 10.1016/j.parkreldis.2016.01.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/30/2015] [Accepted: 01/27/2016] [Indexed: 12/17/2022]
Abstract
In the last decade the possible relationship between Parkinson's disease (PD) and peripheral neuropathy (PN) has received increasing attention. Given that PN is quite common in Parkinson's disease, much controversy has arisen on whether it is part of the neurogenerative process itself - actually one of the possible causes - or a complication of levodopa administration. In this article we will discuss the different hypotheses, as well as our perspective on these open issues.
Collapse
Affiliation(s)
- Giovanni Cossu
- Neurology Department, AOB "G. Brotzu" General Hospital, Cagliari Italy.
| | - Maurizio Melis
- Neurology Department, AOB "G. Brotzu" General Hospital, Cagliari Italy
| |
Collapse
|
89
|
Kimber T, Antonini A. Peripheral neuropathy and levodopa therapy in Parkinson disease: novel insights. Eur J Neurol 2015; 23:435-6. [PMID: 26663039 DOI: 10.1111/ene.12912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- T. Kimber
- Neurology Unit Royal Adelaide Hospital and Department of Medicine University of Adelaide North Terrace Adelaide SA Australia
| | - A. Antonini
- Parkinson and Movement Disorders Unit IRCCS Hospital San Camillo Venice Italy
| |
Collapse
|
90
|
Laboratory assessments in the course of Parkinson’s disease: a clinician’s perspective. J Neural Transm (Vienna) 2015; 123:65-71. [DOI: 10.1007/s00702-015-1481-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 11/02/2015] [Indexed: 12/29/2022]
|
91
|
Merola A, Romagnolo A, Zibetti M, Bernardini A, Cocito D, Lopiano L. Peripheral neuropathy associated with levodopa–carbidopa intestinal infusion: a long‐term prospective assessment. Eur J Neurol 2015; 23:501-9. [DOI: 10.1111/ene.12846] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/04/2015] [Indexed: 11/28/2022]
Affiliation(s)
- A. Merola
- Department of Neuroscience University of Turin Torino Italy
| | - A. Romagnolo
- Department of Neuroscience University of Turin Torino Italy
| | - M. Zibetti
- Department of Neuroscience University of Turin Torino Italy
| | - A. Bernardini
- Department of Neuroscience University of Turin Torino Italy
| | - D. Cocito
- Department of Neuroscience University of Turin Torino Italy
| | - L. Lopiano
- Department of Neuroscience University of Turin Torino Italy
| |
Collapse
|
92
|
Vital A, Lepreux S, Vital C. Peripheral neuropathy and parkinsonism: a large clinical and pathogenic spectrum. J Peripher Nerv Syst 2015; 19:333-42. [PMID: 25582874 DOI: 10.1111/jns.12099] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/26/2014] [Accepted: 12/26/2014] [Indexed: 01/08/2023]
Abstract
Peripheral neuropathy (PN) has been reported in idiopathic and hereditary forms of parkinsonism, but the pathogenic mechanisms are unclear and likely heterogeneous. Levodopa-induced vitamin B12 deficiency has been discussed as a causal factor of PN in idiopathic Parkinson's disease, but peripheral nervous system involvement might also be a consequence of the underlying neurodegenerative process. Occurrence of PN with parkinsonism has been associated with a panel of mitochondrial cytopathies, more frequently related to a nuclear gene defect and mainly polymerase gamma (POLG1) gene. Parkin (PARK2) gene mutations are responsible for juvenile parkinsonism, and possible peripheral nervous system involvement has been reported. Rarely, an association of parkinsonism with PN may be encountered in other neurodegenerative diseases such as fragile X-associated tremor and ataxia syndrome related to premutation CGG repeat expansion in the fragile X mental retardation (FMR1) gene, Machado-Joseph disease related to an abnormal CAG repeat expansion in ataxin-3 (ATXN3) gene, Kufor-Rakeb syndrome caused by mutations in ATP13A2 gene, or in hereditary systemic disorders such as Gaucher disease due to mutations in the β-glucocerebrosidase (GBA) gene and Chediak-Higashi syndrome due to LYST gene mutations. This article reviews conditions in which PN may coexist with parkinsonism.
Collapse
Affiliation(s)
- Anne Vital
- University of Bordeaux, Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, Bordeaux, France; Department of Pathology, Bordeaux University Hospital, Bordeaux, France
| | | | | |
Collapse
|
93
|
Schindlbeck KA, Mehl A, Geffe S, Benik S, Tütüncü S, Klostermann F, Marzinzik F. Somatosensory symptoms in unmedicated de novo patients with idiopathic Parkinson’s disease. J Neural Transm (Vienna) 2015; 123:211-7. [DOI: 10.1007/s00702-015-1459-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/11/2015] [Indexed: 11/28/2022]
|
94
|
Shiga T, Kawata T, Furusho T, Tadokoro T, Suzuki T, Yamamoto Y. Elevation of urinary methylmolonic acid induces the suppression of megalin-mediated endocytotic cycles during vitamin B12 deficiency. Biochem Biophys Res Commun 2015; 465:206-12. [PMID: 26248135 DOI: 10.1016/j.bbrc.2015.07.151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 01/01/2023]
Abstract
Megalin is a scavenger receptor that serves in the endocytosis of a highly diverse group of ligands that includes Vitamin B12. We found an accumulation of megalin closed to apical region in renal proximal tubule cells of Vitamin B12-deficient rats. Interestingly, Vitamin B12 levels also controlled resorption of renal retinol binding protein. Using L2 yolk sac cells, megalin localized to the submembrane compartment by methylmalonic acid (MMA), which accumulates during vitamin B12 deficiency. In addition, MMA inhibited megalin-mediated endocytosis via YWTD repeats motif in an ectodomain of megalin. Therefore, megalin endocytosis may be regulated by MMA.
Collapse
Affiliation(s)
- Takahiro Shiga
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Tetsunori Kawata
- Division of Life, Health, and Sports Education, Graduate School of Education, Okayama University, Okayama, Japan
| | - Tadasu Furusho
- Department of Nutrition, Junior College of Tokyo University of Agriculture, Tokyo, Japan
| | - Tadahiro Tadokoro
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Tsukasa Suzuki
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuji Yamamoto
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
95
|
Zhang L, Yuan Y, Tong Q, Jiang S, Xu Q, Ding J, Zhang L, Zhang R, Zhang K. Reduced plasma taurine level in Parkinson's disease: association with motor severity and levodopa treatment. Int J Neurosci 2015; 126:630-6. [PMID: 26004911 DOI: 10.3109/00207454.2015.1051046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE This study aimed to evaluate the level of taurine in plasma, and its association with the severity of motor and non-motor symptoms (NMS) and chronic levodopa treatment in Parkinson's disease (PD). PATIENTS AND METHODS Plasma taurine level was measured in treated PD (tPD), untreated PD (ntPD) and control groups. Motor symptoms and NMS were assessed using the Unified Parkinson's Disease Rating Scale, the short form of the McGill Pain Questionnaire, the Hamilton Depression Scale, the Scale for Outcomes in Parkinson's disease for Autonomic Symptoms and the Pittsburgh Sleep Quality Index. Longtime exposure to levodopa was indicated by its approximate cumulative dosage. RESULTS The plasma taurine levels of PD patients were decreased when compared with controls and negatively associated with motor severity but not NMS. Moreover, tPD patients exhibited lower levels of plasma taurine than ntPD patients. Interestingly, plasma taurine levels negatively correlated with cumulative levodopa dosage in tPD. After controlling for potential confounders, the association between taurine and levodopa remained significant. CONCLUSION Our study supports that taurine may play important roles in the pathophysiology of PD and the disturbances caused by chronic levodopa administration.
Collapse
Affiliation(s)
- Li Zhang
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Yongsheng Yuan
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Qing Tong
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Siming Jiang
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Qinrong Xu
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Jian Ding
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Lian Zhang
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| | - Rui Zhang
- b 2 Department of Neurosurgery , Nanjing Children's Hospital of Nanjing Medical University , Nanjing , China
| | - Kezhong Zhang
- a 1 Department of Neurology , The First Affiliated Hospital of Nanjing Medical University , Nanjing , China
| |
Collapse
|
96
|
Mylius V, Ciampi de Andrade D, Cury RG, Teepker M, Ehrt U, Eggert KM, Beer S, Kesselring J, Stamelou M, Oertel WH, Möller JC, Lefaucheur JP. Pain in Parkinson's Disease: Current Concepts and a New Diagnostic Algorithm. Mov Disord Clin Pract 2015; 2:357-364. [PMID: 30363602 DOI: 10.1002/mdc3.12217] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 12/30/2022] Open
Abstract
Background Pain is a significant burden for patients with Parkinson's disease (PD) with a high impact on quality of life. The present article aims at summarizing epidemiological, pathophysiological, clinical, and neurophysiological data regarding pain in PD. Methods In this domain, a procedure of systematic assessment is still lacking for the syndromic diagnosis and should take into account pain characteristics, effects of dopaminergic treatment, motor fluctuations, and non-PD-associated pain. Findings We propose an original questionnaire addressing an algorithm suitable for daily clinical practice. The questionnaire is based on a three-step approach addressing first the relationship between pain and PD (including temporal relationship with the course of the disease, association with motor fluctuations, and impact of antiparkinsonian treatment), before classifying pain into one of three main syndromes (i.e., musculoskeletal pain, psychomotor restlessness pain, and neuropathic pain). Conclusions The proposed questionnaire allows the characteristics of each pain type to be determined according to its relationship with the disease and its treatment. The validation of the clinical use of this questionnaire will be the goal of a forthcoming work.
Collapse
Affiliation(s)
- Veit Mylius
- Department of Neurology Philipps University Marburg Germany.,Department of Neurology Center for Neurorehabilitation Valens Switzerland
| | | | - Rubens Gisbert Cury
- Pain Center Department of Neurology University of São Paulo São Paulo SP Brazil
| | | | - Uwe Ehrt
- Psychiatric Clinic Fachklinikum Bernburg Bernburg Germany
| | | | - Serafin Beer
- Department of Neurology Center for Neurorehabilitation Valens Switzerland
| | - Jürg Kesselring
- Department of Neurology Center for Neurorehabilitation Valens Switzerland
| | - Maria Stamelou
- Department of Neurology Philipps University Marburg Germany.,Movement Disorders Clinic Second Department of Neurology University of Athens Athens Greece
| | | | - Jens Carsten Möller
- Parkinson Center Center for Neurological Rehabilitation Zihlschlacht Switzerland
| | - Jean-Pascal Lefaucheur
- Faculté de Médecine Université Paris Est Créteil Créteil France.,Service de Physiologie-Explorations Fonctionnelles Hôpital Henri Mondor Assistance Publique-Hôpitaux de Paris Créteil France
| |
Collapse
|
97
|
Abstract
Electrophysiological examination can provide valuable information on functional abnormalities in patients with Parkinson's disease (PD). Although there are numerous reports on biochemical and molecular alterations in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced experimental parkinsonism in mice, the mode of electrophysiology in this animal model of PD is not clear. This study provides a comparative evaluation of corticomotor evoked potential (CMEP), compound muscle action potential (CMAP) and motor nerve conduction velocity (NCV) in mice treated with MPTP (30 mg/kg, ip, daily for 4 days) or saline (control group). Although the CMEP latencies were similar in both the groups, the CMEP amplitude was non-significantly decreased in MPTP-treated mice. There was a significant increase in the CMAP latency (1.37 ± 0.03 versus 1.20 ± 0.02 ms) and decrease in CMAP amplitude (4.50 ± 0.89 versus 8.31 ± 0.86 mV) in MPTP-treated mice as compared with control group. This prolonged conduction time resulted in a significant decrease in NCV in MPTP-treated mice (21.98 ± 0.54 m/s) as compared with control mice (24.47 ± 0.33 m/s). There was a significant depletion of striatal dopamine in MPTP-treated animals. These findings demonstrate that systemic administration of MPTP significantly impairs both the central and peripheral nervous systems in mice. However, the resemblance of this neurophysiological status with idiopathic PD or other animal models of PD is not clear and requires additional studies.
Collapse
Affiliation(s)
- Haseeb Ahmad Khan
- Analytical and Molecular Bioscience Research Group, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
98
|
Uncini A, Eleopra R, Onofrj M. Polyneuropathy associated with duodenal infusion of levodopa in Parkinson's disease: features, pathogenesis and management. J Neurol Neurosurg Psychiatry 2015; 86:490-5. [PMID: 25168395 DOI: 10.1136/jnnp-2014-308586] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/03/2014] [Indexed: 11/04/2022]
Abstract
Patients with Parkinson's disease (PD) treated with oral levodopa have a higher prevalence of chronic, prevalently sensory, usually mild axonal polyneuropathy (PNP). Several studies showed a positive association among PNP, cumulative levodopa dosage, low serum B12 and high-homocysteine and methylmalonic acid level. Anecdotal severe acute or subacute PNPs thought to be Guillain-Barré syndrome have been reported in patients receiving continuous intraduodenal infusion of levodopa/carbidopa intestinal gel (LCIG). We report an additional acute case and by a systematic literature search we also reviewed the clinical and laboratory features of 13 other acute and 21 subacute PNP cases occurring during LCIG treatment. In series with at least nine patients, the mean frequency of acute and subacute PNP is 13.6% and the mortality rate at 6 months in acute cases is 14%. The great majority of PNP cases displayed axonal sensory-motor and reduced vitamin B12 levels, and alterations of metabolites of 1-carbon pathway were found in most patients. We discuss the possible role of high-levodopa dosage, vitamin B12, B6 and folate deficiency and accumulation of homocysteine and methylmalonic acid in the pathogenesis to conclude that there is enough, although circumstantial, evidence that alterations of 1-carbon pathway are implicated also in acute and subacute PNP during LCIG usage. There is no solid proof for a dysimmune pathogenesis and in our opinion acute, subacute and chronic PNP, either after oral levodopa or LCIG, represent a continuum. Finally, we propose recommendations for prevention and management of PNP occurring during LCIG treatment.
Collapse
Affiliation(s)
- Antonino Uncini
- Department of Neuroscience and Imaging, University "G. d'Annunzio", Chieti-Pescara, Italy
| | - Roberto Eleopra
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Marco Onofrj
- Department of Neuroscience and Imaging, University "G. d'Annunzio", Chieti-Pescara, Italy
| |
Collapse
|
99
|
Clinical Utility of Skin Biopsy in Differentiating between Parkinson's Disease and Multiple System Atrophy. PARKINSONS DISEASE 2015; 2015:167038. [PMID: 25945280 PMCID: PMC4402568 DOI: 10.1155/2015/167038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/24/2015] [Accepted: 03/24/2015] [Indexed: 11/18/2022]
Abstract
Background. It is often difficult to differentiate Parkinson's disease (PD) from multiple system atrophy (MSA), especially in their early stages. Objectives. To examine the clinical utility of histopathological analysis of biopsied skin from the chest wall and/or leg in differentiating between the two diseases. Methods. Skin biopsies from the lower leg and/or anterior chest wall were obtained from 38 patients with idiopathic PD (26 treated with levodopa and 12 levodopa-naïve) and 13 age-matched patients with MSA. We sought aggregates of phosphorylated α-synuclein on cutaneous nerve fibers using double fluorescence immunohistochemistry and confocal microscopy and measured intraepidermal nerve fiber density (IENFD). Results. Phosphorylated α-synuclein aggregates were identified on cutaneous nerves in two patients with PD (5.3%) but in none of the patients with MSA, and IENFD was significantly lower in patients with PD when compared to those with MSA. There was no difference in IENFD between levodopa-treated and levodopa-naïve patients with PD. Conclusions. Our findings suggest that an assessment of IENFD in biopsied skin could be a useful means of differentiating between PD and MSA but that detection of α-synuclein aggregates on cutaneous nerves in the distal sites of the body is insufficiently sensitive.
Collapse
|
100
|
Abdel-Salam OME. Drug therapy for Parkinson’s disease: An update. World J Pharmacol 2015; 4:117-143. [DOI: 10.5497/wjp.v4.i1.117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 01/26/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is the most common neurodegenerative movement disorder, affecting about 1% of the population above the age of 65. PD is characterized by a selective degeneration of the dopaminergic neurons of the substantia nigra pars compacta. This results in a marked loss of striatal dopamine and the development of the characteristic features of the disease, i.e., bradykinesia, rest tremor, rigidity, gait abnormalities and postural instability. Other types of neurons/neurotransmitters are also involved in PD, including cholinergic, serotonergic, glutamatergic, adenosine, and GABAergic neurotransmission which might have relevance to the motor, non-motor, neuropsychiatric and cognitive disturbances that occur in the course of the disease. The treatment of PD relies on replacement therapy with levodopa (L-dopa), the precursor of dopamine, in combination with a peripheral decarboxylase inhibitor (carbidopa or benserazide). The effect of L-dopa, however, declines over time together with the development of motor complications especially dyskinesia in a significant proportion of patients within 5 years of therapy. Other drugs include dopamine-receptor-agonists, catechol-O-methyltransferase inhibitors, monoamine oxidase type B (MAO-B) inhibitors, anticholinergics and adjuvant therapy with the antiviral drug and the N-methyl-D-aspartate glutamate receptor antagonist amantadine. Although, these medications can result in substantial improvements in parkinsonian symptoms, especially during the early stages of the disease, they are often not successful in advanced disease. Moreover, dopaminergic cell death continues over time, emphasizing the need for neuroprotective or neuroregenerative therapies. In recent years, research has focused on non-dopaminergic approach such as the use of A2A receptor antagonists: istradefylline and preladenant or the calcium channel antagonist isradipine. Safinamide is a selective and reversible inhibitor of MAO-B, a glutamate receptor inhibitor as well as sodium and calcium channel blocker. Minocycline and pioglitazone are other agents which have been shown to prevent dopaminergic nigral cell loss in animal models of PD. There is also an evidence to suggest a benefit from iron chelation therapy with deferiprone and from the use of antioxidants or mitochondrial function enhancers such as creatine, alpha-lipoic acid, l-carnitine, and coenzyme Q10.
Collapse
|