1
|
Kambey PA, Wu J, Liu W, Su M, Buberwa W, Tang C. Targeting serum response factor (SRF) deactivates ΔFosB and mitigates Levodopa-induced dyskinesia in a mouse model of Parkinson's disease. Gene Ther 2024; 31:614-624. [PMID: 39384937 DOI: 10.1038/s41434-024-00492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) is currently the preferred treatment for Parkinson's Disease (PD) and is considered the gold standard. However, prolonged use of L-DOPA in patients can result in involuntary movements known as Levodopa-induced dyskinesia (LID), which includes uncontrollable dystonia affecting the trunk, limbs, and face. The role of ΔFosB protein, a truncated splice variant of the FosB gene, in LID has been acknowledged, but its underlying mechanism has remained elusive. Here, using a mouse model of Parkinson's disease treated with chronic levodopa we demonstrate that serum response factor (SRF) binds to the FosB promoter, thereby activating FosB expression and levodopa induced-dyskinetic movements. Western blot analysis demonstrates a significant increase in SRF expression in the dyskinetic group compared to the control group. Knocking down SRF significantly reduced abnormal involuntary movements (AIMS) and ΔFosB expression compared to the control. Conversely, overexpression of SRF led to an increase in ΔFosB expression and worsened levodopa-induced dyskinesia. To shed light on the regulatory role of the Akt signaling pathway in this phenomenon, we administered the Akt agonist SC79 to PD mouse models via intraperitoneal injection, followed by L-DOPA administration. The expression of SRF, ΔFosB, and phosphorylated Akt (p-Akt) significantly increased in this group compared to the group receiving normal saline to signify that these happen through Akt signaling pathway. Collectively, our findings identify a promising therapeutic target for addressing levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Huangpu District, Guangzhou, China.
| | - Jiao Wu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingyu Su
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wokuheleza Buberwa
- Department of Neurology, The second affiliated hospital of Xi'an Jiaotong University, 710049, Xi'an, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
2
|
Nomoto M, Tsuboi Y, Kashihara K, Chiu SW, Maeda T, Saiki H, Watanabe H, Shimo Y, Hattori N, Yamaguchi T. Prescription trends in Japanese advanced Parkinson's disease patients with non-motor symptoms: J-FIRST. PLoS One 2024; 19:e0309297. [PMID: 39441810 PMCID: PMC11498663 DOI: 10.1371/journal.pone.0309297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/01/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Non-motor symptoms (NMS) are important factors when selecting treatments for patients with advanced Parkinson's disease (PD). We sought to elucidate the prescribing practices for advanced PD patients with NMS in Japanese clinical practice. METHODS We examined the prescription rates and doses of anti-PD drugs, and the use of non-steroidal anti-inflammatory drugs (NSAIDs) in post hoc analyses of a 52-week observational study of 996 PD patients with wearing-off on levodopa-containing therapy and ≥1 NMS. RESULTS Dopamine agonists were the most frequently prescribed drugs combined with levodopa-containing drugs, followed by entacapone, zonisamide, istradefylline, selegiline, and amantadine. The daily dose of levodopa-containing drugs, rotigotine, entacapone, istradefylline, and droxidopa, and the levodopa-equivalent dose increased during the observation period. In a subgroup analysis of patients stratified by NMS status (improved/unchanged/deteriorated), the deteriorated group had higher prescription rates of entacapone and istradefylline, whereas the improved group had higher prescription rates of NSAIDs and zonisamide at Week 52. Prescriptions varied by geographical region for anti-PD drugs and by NMS status for NSAIDs. CONCLUSIONS There were significant changes in the prescriptions and dosing of selected anti-PD drugs, especially newer drugs. Anti-PD drug and NSAID prescriptions also varied by changes in NMS status and geographic region.
Collapse
Affiliation(s)
- Masahiro Nomoto
- Department of Neurology and Clinical Pharmacology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | | | - Shih-Wei Chiu
- Division of Biostatistics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tetsuya Maeda
- Department of Neurology, Research Institute for Brain and Blood Vessels-Akita, Akita, Japan
| | - Hidemoto Saiki
- Department of Neurology, Kitano Hospital, The Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | | | - Yasushi Shimo
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takuhiro Yamaguchi
- Division of Biostatistics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | |
Collapse
|
3
|
Zhang Y, Zhu XB, Zhao Y, Cui GY, Li WT, Yuan CX, Huang JP, Wan Y, Wu N, Song L, Zhao JH, Liang Y, Xu CY, Liu MJ, Gao C, Chen XX, Liu ZG. Efficacy and safety of Tianqi Pingchan Granule, a compound Chinese herbal medicine, for levodopa-induced dyskinesia in Parkinson's disease: A randomized double-blind placebo-controlled trial. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:545-551. [PMID: 39060125 DOI: 10.1016/j.joim.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 04/15/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Patients with Parkinson's disease (PD) undergoing long-term levodopa therapy are prone to develop levodopa-induced dyskinesia (LID). Amantadine is the main drug recommended for the treatment of LID by current guidelines, but it is far from meeting clinical needs. Tianqi Pingchan Granule (TPG), a compound Chinese herbal medicine, has been developed to relieve symptom of LID. OBJECTIVE This randomized controlled trial evaluated the efficacy and safety of the combination of TPG and amantadine for LID. DESIGN, SETTING, PARTICIPANTS AND INTERVENTIONS This is a randomized double-blind placebo-controlled trial, conducted from January 2020 to August 2021 at 6 sites in Jiangsu, Zhejiang and Shanghai, China. One hundred PD patients with ≥ 0.5 h of LID were randomly assigned to either the TPG plus amantadine group (TPG group) or the placebo plus amantadine group (placebo group), and treated for a period of 12 weeks. To ensure unbiased results, all study participants, investigators and sponsors were unaware of group allocations. Additionally, the data analysts remained blinded until the analysis was finalized. MAIN OUTCOME MEASURES The primary outcome was assessed using the Unified Dyskinesia Rating Scale (UDysRS) (Range 0-104). The key secondary end point was improvement of motor and non-motor symptoms. Safety analyses included all enrolled patients. RESULTS One hundred patients were enrolled and randomized into the two treatment groups. The changes in UDysRS at week 12 were -11.02 for the TPG group and -4.19 for the placebo group (treatment difference -6.83 [-10.53 to -3.12]; P = 0.0004). Adverse events were reported for 2 of 50 patients (4.0%) in each of the groups. CONCLUSION This study indicated that a 12-week treatment of amantadine plus TPG effectively reduced UDysRS scores and was well tolerated, demonstrating the efficacy and safety of TPG for the treatment of LID in PD. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT04173832. PLEASE CITE THIS ARTICLE AS Zhang Y, Zhu XB, Zhao Y, Cui GY, Li WT, Yuan CX, Huang JP, Wan Y, Wu N, Song L, Zhao JH, Liang Y, Xu CY, Liu MJ, Gao C, Chen XX, Liu ZG. Efficacy and safety of Tianqi Pingchan Granule, a compound Chinese herbal medicine, for levodopa-induced dyskinesia in Parkinson's disease: A randomized double-blind placebo-controlled trial. J Integr Med. 2024; 22(5): 545-551.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiao-Bo Zhu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Gui-Yun Cui
- Parkinson's Disease Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Wen-Tao Li
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Can-Xing Yuan
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jian-Ping Huang
- Department of Neurology, Wenzhou Central Hospital, Wenzhou 325000, Zhejiang Province, China
| | - Ying Wan
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Na Wu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lu Song
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jia-Hao Zhao
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Liang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Chuan-Ying Xu
- Parkinson's Disease Center, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Mei-Juan Liu
- Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Chen Gao
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xin-Xin Chen
- Department of Neurology, Wenzhou Central Hospital, Wenzhou 325000, Zhejiang Province, China
| | - Zhen-Guo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
4
|
Yoo HS, Kim HK, Lee HS, Yoon SH, Na HK, Kang SW, Lee JH, Ryu YH, Lyoo CH. Predictors associated with the rate of progression of nigrostriatal degeneration in Parkinson's disease. J Neurol 2024; 271:5213-5222. [PMID: 38839638 DOI: 10.1007/s00415-024-12477-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/20/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Parkinson's disease (PD) manifests as a wide variety of clinical phenotypes and its progression varies greatly. However, the factors associated with different disease progression remain largely unknown. METHODS In this retrospective cohort study, we enrolled 113 patients who underwent 18F-FP-CIT PET scan twice. Given the negative exponential progression pattern of dopamine loss in PD, we applied the natural logarithm to the specific binding ratio (SBR) of two consecutive 18F-FP-CIT PET scans and conducted linear mixed model to calculate individual slope to define the progression rate of nigrostriatal degeneration. We investigated the clinical and dopamine transporter (DAT) availability patterns associated with the progression rate of dopamine depletion in each striatal sub-region. RESULTS More symmetric parkinsonism, the presence of dyslipidemia, lower K-MMSE total score, and lower anteroposterior gradient of the mean putaminal SBR were associated with faster progression rate of dopamine depletion in the caudate nucleus. More symmetric parkinsonism and lower anteroposterior gradient of the mean putaminal SBR were associated with faster depletion of dopamine in the anterior putamen. Older age at onset, more symmetric parkinsonism, the presence of dyslipidemia, and lower anteroposterior gradient of the mean putaminal SBR were associated with faster progression rate of dopamine depletion in the posterior putamen. Lower striatal mean SBR predicted the development of LID, while lower mean SBR in the caudate nuclei predicted the development of dementia. DISCUSSION Our results suggest that the evaluation of baseline clinical features and patterns of DAT availability can predict the progression of PD and its prognosis.
Collapse
Affiliation(s)
- Han Soo Yoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Han-Kyeol Kim
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - So Hoon Yoon
- Department of Neurology, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, South Korea
| | - Han Kyu Na
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Sung Woo Kang
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Jae-Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea.
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea.
| |
Collapse
|
5
|
Postuma RB, Weintraub D, Simuni T, Rodríguez‐Violante M, Leentjens AF, Hu MT, Espay AJ, Erro R, Dujardin K, Bohnen NI, Berg D, Mestre TA, Marras C. Anticipating Tomorrow: Tailoring Parkinson's Symptomatic Therapy Using Predictors of Outcome. Mov Disord Clin Pract 2024; 11:983-991. [PMID: 38817000 PMCID: PMC11329576 DOI: 10.1002/mdc3.14089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Although research into Parkinson's disease (PD) subtypes and outcome predictions has continued to advance, recommendations for using outcome prediction to guide current treatment decisions remain sparse. OBJECTIVES To provide expert opinion-based recommendations for individually tailored PD symptomatic treatment based on knowledge of risk prediction and subtypes. METHODS Using a modified Delphi approach, members of the Movement Disorders Society (MDS) Task Force on PD subtypes generated a series of general recommendations around the question: "Using what you know about genetic/biological/clinical subtypes (or any individual-level predictors of outcome), what advice would you give for selecting symptomatic treatments for an individual patient now, based on what their subtype or individual characteristics predict about their future disease course?" After four iterations and revisions, those recommendations with over 75% endorsement were adopted. RESULTS A total of 19 recommendations were endorsed by a group of 13 panelists. The recommendations primarily centered around two themes: (1) incorporating future risk of cognitive impairment into current treatment plans; and (2) identifying future symptom clusters that might be forestalled with a single medication. CONCLUSIONS These recommendations provide clinicians with a framework for integrating future outcomes into patient-specific treatment choices. They are not prescriptive guidelines, but adaptable suggestions, which should be tailored to each individual. They are to be considered as a first step of a process that will continue to evolve as additional stakeholders provide new insights and as new information becomes available. As individualized risk prediction advances, the path to better tailored treatment regimens will become clearer.
Collapse
Affiliation(s)
- Ronald B. Postuma
- Department of NeurologyMontreal Neurological Institute, McGill UniversityMontrealQuebecCanada
| | - Daniel Weintraub
- Departments of Psychiatry and Neurology, Perelman School of Medicine at the University of Pennsylvania; Parkinson's Disease Research, Education and Clinical Center (PADRECC)Philadelphia Veterans Affairs Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Tanya Simuni
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | | | - Albert F.G. Leentjens
- Department of PsychiatryMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Michele T. Hu
- Nuffield Department of Clinical Neurosciences, Neurology DepartmentOxford University and John Radcliffe HospitalOxfordUnited Kingdom
| | - Alberto J. Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of NeurologyUniversity of CincinnatiCincinnatiOhioUSA
| | - Roberto Erro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Neuroscience SectionUniversity of SalernoBaronissiItaly
| | - Kathy Dujardin
- Neurology and Movement Disorders DepartmentUniversity of Lille, Inserm, Lille Neurosciences and Cognition, CHU‐LilleLilleFrance
| | - Nicolaas I. Bohnen
- Departments of Radiology and NeurologyUniversity of Michigan, University of Michigan Udall Center, Ann Arbor VAMCAnn ArborMichiganUSA
| | - Daniela Berg
- Department of NeurologyChristian‐Albrechts‐UniversityKielGermany
| | - Tiago A. Mestre
- Division of Neurology, Department of MedicineUniversity of Ottawa, The University of Ottawa Brain and Research InstituteOttawaOntarioCanada
- Parkinson's Disease and Movement Disorders ClinicThe Ottawa Hospital, The Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Connie Marras
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders ClinicToronto Western Hospital, University Health NetworkTorontoOntarioCanada
| |
Collapse
|
6
|
Marano M, Pilotto A, Padovani A, Gupta D, Vivacqua G, Magliozzi A, Di Lazzaro V, Carta M, Meloni M. The chronic use of serotonin norepinephrine reuptake inhibitors facilitates dyskinesia priming in early Parkinson's disease. J Neurol 2024; 271:3711-3720. [PMID: 38720139 DOI: 10.1007/s00415-024-12400-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/24/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Parkinson's disease (PD) patients are frequently exposed to antidepressant medications (ADMs). Norepinephrine (NE) and serotonin (5HT) systems have a role in levodopa-induced dyskinesias (LID) pathophysiology. METHODS We performed a longitudinal analysis on the PPMI cohort including drug-naïve PD patients, who are progressively exposed to dopamine replacement therapies (DRTs) to test the effect of ADM exposure on LID development by the 4th year of follow-up. RESULTS LID prevalence (according to MDS UPDRS score 4.1 ≥ 1) was 16% (42/251); these patients were more likely women (p = 0.01), had higher motor (p < 0.001) and depression scores (p = 0.01) and lower putaminal DAT binding ratio (p = 0.01). LID were associated with the exposure time to L-DOPA (2.2 ± 1.07 vs 2.6 ± 0.9, p = 0.02) and to the exposure to ADMs, in particular to SNRI (4.8% vs 21.4%, p < 0.001). The latter persisted after correcting for significant covariates (e.g., disease duration, cognitive status, motor impairment, depression, dopaminergic denervation). A similar difference in LID prevalence in PD patients exposed vs non-exposed to SNRI was observed on matched data by the real-world TriNetX repository (22% vs 13%, p < 0.001). DISCUSSION This study supports the presence of an effect of SNRI on LID priming in patients with early PD. Independent prospective cohort studies are warranted to further verify such association.
Collapse
Affiliation(s)
- Massimo Marano
- Neurology, Neurophysiology, Neurobiology and Psychiatry Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128, Rome, Italy.
- Fondazione Policlinico Universitario Campus Bio-Medico, Viale Alvaro del Portillo 200, 00128, Rome, Italy.
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| | - Deepak Gupta
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Giorgio Vivacqua
- Laboratory of Microscopic and Ultrastructural Anatomy, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128, Roma, Italy
| | - Alessandro Magliozzi
- Neurology, Neurophysiology, Neurobiology and Psychiatry Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128, Rome, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Viale Alvaro del Portillo 200, 00128, Rome, Italy
| | - Vincenzo Di Lazzaro
- Neurology, Neurophysiology, Neurobiology and Psychiatry Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128, Rome, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Viale Alvaro del Portillo 200, 00128, Rome, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, CA, Italy
| | - Mario Meloni
- Neurology Unit, Azienda Ospedaliera Universitaria di Cagliari, Cagliari, Italy
| |
Collapse
|
7
|
Santos-García D, de Deus T, Cores C, Feal Painceiras MJ, Íñiguez Alvarado MC, Samaniego LB, López Maside A, Jesús S, Cosgaya M, García Caldentey J, Caballol N, Legarda I, Hernández-Vara J, Cabo López I, López Manzanares L, González-Aramburu I, Ávila A, Gómez-Mayordomo V, Nogueira V, Dotor García-Soto J, Borrué-Fernández C, Solano B, Álvarez Sauco M, Vela L, Escalante S, Cubo E, Mendoza Z, Pareés I, Sánchez Alonso P, Alonso Losada MG, López-Ariztegui N, Gastón I, Kulisevsky J, Seijo M, Valero C, Alonso Redondo R, Buongiorno MT, Ordás C, Menéndez-González M, McAfee D, Martinez-Martin P, Mir P. Levodopa-Induced Dyskinesias are Frequent and Impact Quality of Life in Parkinson's Disease: A 5-Year Follow-Up Study. Mov Disord Clin Pract 2024; 11:830-849. [PMID: 38747234 PMCID: PMC11233927 DOI: 10.1002/mdc3.14056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/19/2024] [Accepted: 03/29/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Levodopa-induced dyskinesias (LID) are frequent in Parkinson's disease (PD). OBJECTIVE To analyze the change in the frequency of LID over time, identify LID related factors, and characterize how LID impact on patients' quality of life (QoL). PATIENTS AND METHODS PD patients from the 5-year follow-up COPPADIS cohort were included. LID were defined as a non-zero score in the item "Time spent with dyskinesia" of the Unified Parkinson's Disease Rating Scale-part IV (UPDRS-IV). The UPDRS-IV was applied at baseline (V0) and annually for 5 years. The 39-item Parkinson's disease Questionnaire Summary Index (PQ-39SI) was used to asses QoL. RESULTS The frequency of LID at V0 in 672 PD patients (62.4 ± 8.9 years old; 60.1% males) with a mean disease duration of 5.5 ± 4.3 years was 18.9% (127/672) and increased progressively to 42.6% (185/434) at 5-year follow-up (V5). The frequency of disabling LID, painful LID, and morning dystonia increased from 6.9%, 3.3%, and 10.6% at V0 to 17.3%, 5.5%, and 24% at V5, respectively. Significant independent factors associated with LID (P < 0.05) were a longer disease duration and time under levodopa treatment, a higher dose of levodopa, a lower weight and dose of dopamine agonist, pain severity and the presence of motor fluctuations. LID at V0 (β = 0.073; P = 0.027; R2 = 0.62) and to develop disabling LID at V5 (β = 0.088; P = 0.009; R2 = 0.73) were independently associated with a higher score on the PDQ-39SI. CONCLUSION LID are frequent in PD patients. A higher dose of levodopa and lower weight were factors associated to LID. LID significantly impact QoL.
Collapse
Affiliation(s)
| | - Teresa de Deus
- CHUF, Complejo Hospitalario Universitario de Ferrol, A Coruña, Spain
| | - Carlos Cores
- CHUAC, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | | | | | - Lucía B Samaniego
- CHUAC, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | | | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Nuria Caballol
- Consorci Sanitari Integral, Hospital Moisés Broggi, Sant Joan Despí, Spain
| | - Ines Legarda
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Jorge Hernández-Vara
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Iria Cabo López
- Complejo Hospitalario Universitario de Pontevedra (CHOP), Pontevedra, Spain
| | | | - Isabel González-Aramburu
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Hospital Universitario Marqués de Valdecilla - IDIVAL, Santander, Spain
| | - Asunción Ávila
- Consorci Sanitari Integral, Hospital General de L'Hospitalet, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Víctor Gómez-Mayordomo
- Neurology Department, Institute of Neuroscience, Vithas Madrid La Milagrosa University Hospital, Vithas Hospital Group, Madrid, Spain
| | | | | | | | - Berta Solano
- Institut d'Assistència Sanitària (IAS) - Institut Català de la Salut, Girona, Spain
| | | | - Lydia Vela
- Fundación Hospital de Alcorcón, Madrid, Spain
| | - Sonia Escalante
- Hospital de Tortosa Verge de la Cinta (HTVC), Tortosa, Spain
| | - Esther Cubo
- Complejo Asistencial Universitario de Burgos, Burgos, Spain
| | - Zebenzui Mendoza
- Hospital Universitario de Canarias, San Cristóbal de la Laguna, Santa Cruz de Tenerife, Spain
| | - Isabel Pareés
- Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | | | - Maria G Alonso Losada
- Hospital Álvaro Cunqueiro, Complejo Hospitalario Universitario de Vigo (CHUVI), Vigo, Spain
| | | | | | - Jaime Kulisevsky
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Hospital de Sant Pau, Barcelona, Spain
| | - Manuel Seijo
- Complejo Hospitalario Universitario de Pontevedra (CHOP), Pontevedra, Spain
| | | | | | | | | | | | - Darrian McAfee
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pablo Martinez-Martin
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, 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/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
8
|
Gandhi SE, Zerenner T, Nodehi A, Lawton MA, Marshall V, Al‐Hajraf F, Grosset KA, Morris HR, Hu MT, Ben‐Shlomo Y, Grosset DG. Motor Complications in Parkinson's Disease: Results from 3343 Patients Followed for up to 12 Years. Mov Disord Clin Pract 2024; 11:686-697. [PMID: 38587023 PMCID: PMC11145112 DOI: 10.1002/mdc3.14044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/26/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Motor complications are well recognized in Parkinson's disease (PD), but their reported prevalence varies and functional impact has not been well studied. OBJECTIVES To quantify the presence, severity, impact and associated factors for motor complications in PD. METHODS Analysis of three large prospective cohort studies of recent-onset PD patients followed for up to 12 years. The MDS-UPDRS part 4 assessed motor complications and multivariable logistic regression tested for associations. Genetic risk score (GRS) for Parkinson's was calculated from 79 single nucleotide polymorphisms. RESULTS 3343 cases were included (64.7% male). Off periods affected 35.0% (95% CI 33.0, 37.0) at 4-6 years and 59.0% (55.6, 62.3) at 8-10 years. Dyskinesia affected 18.5% (95% CI 16.9, 20.2) at 4-6 years and 42.1% (38.7, 45.5) at 8-10 years. Dystonia affected 13.4% (12.1, 14.9) at 4-6 years and 22.8% (20.1, 25.9) at 8-10 years. Off periods consistently caused greater functional impact than dyskinesia. Motor complications were more common among those with higher drug doses, younger age at diagnosis, female gender, and greater dopaminergic responsiveness (in challenge tests), with associations emerging 2-4 years post-diagnosis. Higher Parkinson's GRS was associated with early dyskinesia (0.026 ≤ P ≤ 0.050 from 2 to 6 years). CONCLUSIONS Off periods are more common and cause greater functional impairment than dyskinesia. We confirm previously reported associations between motor complications with several demographic and medication factors. Greater dopaminergic responsiveness and a higher genetic risk score are two novel and significant independent risk factors for the development of motor complications.
Collapse
Affiliation(s)
- Sacha E. Gandhi
- School of Neuroscience and PsychologyUniversity of GlasgowGlasgowUnited Kingdom
| | - Tanja Zerenner
- Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Anahita Nodehi
- Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Michael A. Lawton
- Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | | | - Falah Al‐Hajraf
- Oxford Parkinson's Disease Centre, Nuffield Department of Clinical NeuroscienceOxford UniversityOxfordUnited Kingdom
- Department of Pharmacology and Toxicology, Faculty of MedicineKuwait UniversityKuwait CityKuwait
| | | | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUnited Kingdom
| | - Michele T. Hu
- Oxford Parkinson's Disease Centre, Nuffield Department of Clinical NeuroscienceOxford UniversityOxfordUnited Kingdom
| | - Yoav Ben‐Shlomo
- Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Donald G. Grosset
- School of Neuroscience and PsychologyUniversity of GlasgowGlasgowUnited Kingdom
| |
Collapse
|
9
|
Mulroy E, Erro R, Bhatia KP, Hallett M. Refining the clinical diagnosis of Parkinson's disease. Parkinsonism Relat Disord 2024; 122:106041. [PMID: 38360507 PMCID: PMC11069446 DOI: 10.1016/j.parkreldis.2024.106041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Our ability to define, understand, and classify Parkinson's disease (PD) has undergone significant changes since the disorder was first described in 1817. Clinical features and neuropathologic signatures can now be supplemented by in-vivo interrogation of genetic and biological substrates of disease, offering great opportunity for further refining the diagnosis of PD. In this mini-review, we discuss the historical perspectives which shaped our thinking surrounding the definition and diagnosis of PD. We highlight the clinical, genetic, pathologic and biologic diversity which underpins the condition, and proceed to discuss how recent developments in our ability to define biologic and pathologic substrates of disease might impact PD definition, diagnosis, individualised prognostication, and personalised clinical care. We argue that Parkinson's 'disease', as currently diagnosed in the clinic, is actually a syndrome. It is the outward manifestation of any array of potential dysfunctional biologic processes, neuropathological changes, and disease aetiologies, which culminate in common outward clinical features which we term PD; each person has their own unique disease, which we can now define with increasing precision. This is an exciting time in PD research and clinical care. Our ability to refine the clinical diagnosis of PD, incorporating in-vivo assessments of disease biology, neuropathology, and neurogenetics may well herald the era of biologically-based, precision medicine approaches PD management. With this however comes a number of challenges, including how to integrate these technologies into clinical practice in a way which is acceptable to patients, promotes meaningful changes to care, and minimises health economic impact.
Collapse
Affiliation(s)
- Eoin Mulroy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Roberto Erro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, (SA), Italy
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
10
|
Zhang Y, Zhu XB, Gan J, Song L, Qi C, Wu N, Wan Y, Hou M, Liu Z. Impulse control behaviors and apathy commonly co-occur in de novo Parkinson's disease and predict the incidence of levodopa-induced dyskinesia. J Affect Disord 2024; 351:895-903. [PMID: 38342317 DOI: 10.1016/j.jad.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
OBJECTIVE Impulse control behaviors (ICBs) and apathy are believed to represent opposite motivational expressions of the same behavioral spectrum involving hypo- and hyperdopaminergic status, but this has been recently debated. Our study aims to estimate the co-occurrence of ICBs and apathy in early Parkinson's disease (PD) and to determine whether this complex neuropsychiatric condition is an important marker of PD prognoses. METHODS Neuropsychiatric symptoms, clinical data, neuroimaging results, and demographic data from de novo PD patients were obtained from the Parkinson's Progression Markers Initiative, a prospective, multicenter, observational cohort. The clinical characteristics of ICBs co-occurring with apathy and their prevalence were analyzed. We compared the prognoses of the different groups during the 8-year follow-up. Multivariate Cox regression analysis was conducted to predict the development of levodopa-induced dyskinesia (LID) using baseline neuropsychiatric symptoms. RESULTS A total of 422 PD patients and 195 healthy controls (HCs) were included. In brief, 87 (20.6 %) de novo PD patients and 37 (19.0 %) HCs had ICBs at baseline. Among them, 23 (26.4 %) de novo PD patients and 3 (8.1 %) HCs had clinical symptoms of both ICBs and apathy. The ICBs and apathy group had more severe non-motor symptoms than the isolated ICBs group. Cox regression analysis demonstrated that the co-occurrence of ICBs and apathy was a risk factor for LID development (HR 2.229, 95 % CI 1.209 to 4.110, p = 0.010). CONCLUSIONS Co-occurrence of ICBs and apathy is common in patients with early PD and may help to identify the risk of LID development.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Xiao Bo Zhu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China; Department of Neurology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, 1158 Gong yuan East Road, Shanghai 201700, People's Republic of China
| | - Jing Gan
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Lu Song
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Chen Qi
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Na Wu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Ying Wan
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Miaomiao Hou
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kong jiang Road, Shanghai 200092, People's Republic of China.
| |
Collapse
|
11
|
Riasi A, Delrobaei M, Salari M. A decision support system based on recurrent neural networks to predict medication dosage for patients with Parkinson's disease. Sci Rep 2024; 14:8424. [PMID: 38600209 PMCID: PMC11006681 DOI: 10.1038/s41598-024-59179-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 04/08/2024] [Indexed: 04/12/2024] Open
Abstract
Using deep learning has demonstrated significant potential in making informed decisions based on clinical evidence. In this study, we deal with optimizing medication and quantitatively present the role of deep learning in predicting the medication dosage for patients with Parkinson's disease (PD). The proposed method is based on recurrent neural networks (RNNs) and tries to predict the dosage of five critical medication types for PD, including levodopa, dopamine agonists, monoamine oxidase-B inhibitors, catechol-O-methyltransferase inhibitors, and amantadine. Recurrent neural networks have memory blocks that retain crucial information from previous patient visits. This feature is helpful for patients with PD, as the neurologist can refer to the patient's previous state and the prescribed medication to make informed decisions. We employed data from the Parkinson's Progression Markers Initiative. The dataset included information on the Unified Parkinson's Disease Rating Scale, Activities of Daily Living, Hoehn and Yahr scale, demographic details, and medication use logs for each patient. We evaluated several models, such as multi-layer perceptron (MLP), Simple-RNN, long short-term memory (LSTM), and gated recurrent units (GRU). Our analysis found that recurrent neural networks (LSTM and GRU) performed the best. More specifically, when using LSTM, we were able to predict levodopa and dopamine agonist dosage with a mean squared error of 0.009 and 0.003, mean absolute error of 0.062 and 0.030, root mean square error of 0.099 and 0.053, and R-squared of 0.514 and 0.711, respectively.
Collapse
Affiliation(s)
- Atiye Riasi
- Department of Biomedical Engineering, Faculty of Electrical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Mehdi Delrobaei
- Department of Mechatronics, Faculty of Electrical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, Western University, London, ON, Canada.
| | - Mehri Salari
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Trevisan L, Gaudio A, Monfrini E, Avanzino L, Di Fonzo A, Mandich P. Genetics in Parkinson's disease, state-of-the-art and future perspectives. Br Med Bull 2024; 149:60-71. [PMID: 38282031 PMCID: PMC10938543 DOI: 10.1093/bmb/ldad035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disorder and is clinically characterized by the presence of motor (bradykinesia, rigidity, rest tremor and postural instability) and non-motor symptoms (cognitive impairment, autonomic dysfunction, sleep disorders, depression and hyposmia). The aetiology of PD is unknown except for a small but significant contribution of monogenic forms. SOURCES OF DATA No new data were generated or analyzed in support of this review. AREAS OF AGREEMENT Up to 15% of PD patients carry pathogenic variants in PD-associated genes. Some of these genes are associated with mendelian inheritance, while others act as risk factors. Genetic background influences age of onset, disease course, prognosis and therapeutic response. AREAS OF CONTROVERSY Genetic testing is not routinely offered in the clinical setting, but it may have relevant implications, especially in terms of prognosis, response to therapies and inclusion in clinical trials. Widely adopted clinical guidelines on genetic testing are still lacking and open to debate. Some new genetic associations are still awaiting confirmation, and selecting the appropriate genes to be included in diagnostic panels represents a difficult task. Finally, it is still under study whether (and to which degree) specific genetic forms may influence the outcome of PD therapies. GROWING POINTS Polygenic Risk Scores (PRS) may represent a useful tool to genetically stratify the population in terms of disease risk, prognosis and therapeutic outcomes. AREAS TIMELY FOR DEVELOPING RESEARCH The application of PRS and integrated multi-omics in PD promises to improve the personalized care of patients.
Collapse
Affiliation(s)
- L Trevisan
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Largo P. Daneo 3, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino – SS Centro Tumori Ereditari, Largo R. Benzi 10, Genova, 16132, Italy
| | - A Gaudio
- IRCCS Ospedale Policlinico San Martino- UOC Genetica Medica, Largo R. Benzi 10, Genova, 16132, Italy
| | - E Monfrini
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 35, Milan, 20122, Italy
- Neurology Unit, Foundation IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via Festa del Perdono 7, Milan, 20122, Italy
| | - L Avanzino
- Department of Experimental Medicine, Section of Human Physiology, University of Genoa, Viale Benedetto XV/3, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 3, Genova, 16132, Italy
| | - A Di Fonzo
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 35, Milan, 20122, Italy
- Neurology Unit, Foundation IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via Festa del Perdono 7, Milan, 20122, Italy
| | - P Mandich
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Largo P. Daneo 3, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino- UOC Genetica Medica, Largo R. Benzi 10, Genova, 16132, Italy
| |
Collapse
|
13
|
Gan Y, Su D, Zhang Z, Zhang Z, Yan R, Liu Z, Wang Z, Zhou J, Lam JST, Wu T, Jing J, Feng T. Microstructural and functional alterations of the ventral pallidum are associated with levodopa-induced dyskinesia in Parkinson's disease. Eur J Neurol 2024; 31:e16147. [PMID: 37975786 PMCID: PMC11235694 DOI: 10.1111/ene.16147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND PURPOSE The ventral pallidum (VP) regulates involuntary movements, but it is unclear whether the VP regulates the abnormal involuntary movements in Parkinson's disease (PD) patients who have levodopa-induced dyskinesia (LID). To further understand the role of the VP in PD patients with LID (PD-LID), we explored the structural and functional characteristics of the VP in such patients using multimodal magnetic resonance imaging (MRI). METHODS Thirty-one PD-LID patients, 39 PD patients without LID (PD-nLID), and 28 healthy controls (HCs) underwent T1-weighted MRI, quantitative susceptibility mapping, multi-shell diffusion MRI, and resting-state functional MRI (rs-fMRI). Different measures characterizing the VP were obtained using a region-of-interest-based approach. RESULTS The left VP in the PD-LID group showed significantly higher intracellular volume fraction (ICVF) and isotropic volume fraction (IsoVF) compared with the PD-nLID and HC groups. Rs-MRI revealed that, compared with the PD-nLID group, the PD-LID group in the medication 'off' state had higher functional connectivity (FC) between the left VP and the left anterior caudate, left middle frontal gyrus and left precentral gyrus, as well as between the right VP and the right posterior ventral putamen and right mediodorsal thalamus. In addition, the ICVF values of the left VP, the FC between the left VP and the left anterior caudate and left middle frontal gyrus were positively correlated with Unified Dyskinesia Rating Scale scores. CONCLUSION Our multimodal imaging findings show that the microstructural changes of the VP (i.e., the higher ICVF and IsoVF) and the functional change in the ventral striatum-VP-mediodorsal thalamus-cortex network may be associated with pathophysiological mechanisms of PD-LID.
Collapse
Affiliation(s)
- Yawen Gan
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Dongning Su
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Zhe Zhang
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Tiantan Neuroimaging Center of Excellence, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Zhijin Zhang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Rui Yan
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Zhu Liu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Zhan Wang
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Junhong Zhou
- Hinda and Arthur Marcus Institute for Aging ResearchHebrew SeniorLifeRoslindaleMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Joyce S. T. Lam
- Pacific Parkinson's Research Centre, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Tao Wu
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jing Jing
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Tiantan Neuroimaging Center of Excellence, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Tao Feng
- Department of Neurology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
14
|
Sarkar S, Roy A, Choudhury S, Banerjee R, Dey S, Kumar H. Levodopa-induced Dyskinesia in Parkinson's Disease: Plausible Inflammatory and Oxidative Stress Biomarkers. Can J Neurol Sci 2024; 51:104-109. [PMID: 36660782 DOI: 10.1017/cjn.2023.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Pathophysiology of levodopa-induced dyskinesia (LID) remains obscure. Increased dopamine metabolism due to prolonged levodopa treatment can exacerbate oxidative damage and neuroinflammatory pathology in Parkinson's disease (PD). Association of novel peripheral markers with LID severity might provide insight into LID pathomechanisms. OBJECTIVE We aimed to study specific peripheral blood inflammatory-oxidative markers in LID patients and investigate their association with clinical severity of LID. METHOD Motor, non-motor and cognitive changes in PD with and without LID compared to healthy-matched controls were identified. Within the same cohort, inflammatory marker (sLAG3, TOLLIP, NLRP3 and IL-1β) levels and antioxidant enzyme activities were determined by ELISA and spectrophotometric methods. RESULTS LID patients showed distinctly upregulated TOLLIP, IL-1β levels with significant diminution of antioxidant activity compared to controls. Significant negative association of cognitive markers with oxidative changes was also observed. CONCLUSION To our understanding, this is the first study that indicates the involvement of toll-like receptor-mediated distinct and low-grade inflammatory activation in LID pathophysiology.
Collapse
Affiliation(s)
- Swagata Sarkar
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata, India
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Akash Roy
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata, India
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Supriyo Choudhury
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata, India
| | - Rebecca Banerjee
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata, India
| | - Sanjit Dey
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Hrishikesh Kumar
- Department of Neurology, Institute of Neurosciences Kolkata, Kolkata, India
| |
Collapse
|
15
|
Leal DAB, Dias CMV, Ramos RP, Brys I. Prediction of dyskinesia in Parkinson's disease patients using machine learning algorithms. Sci Rep 2023; 13:22426. [PMID: 38104147 PMCID: PMC10725420 DOI: 10.1038/s41598-023-49617-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023] Open
Abstract
Dyskinesias are non preventable abnormal involuntary movements that represent the main challenge of the long term treatment of Parkinson's disease (PD) with the gold standard dopamine precursor levodopa. Applying machine learning techniques on the data extracted from the Parkinson's Progression Marker Initiative (PPMI, Michael J. Fox Foundation), this study was aimed to identify PD patients who are at high risk of developing dyskinesias. Data regarding clinical, behavioral and neurological features from 697 PD patients were included in our study. Our results show that the Random Forest was the classifier with the best and most consistent performance, reaching an area under the receiver operating characteristic (ROC) curve of up to 91.8% with only seven features. Information regarding the severity of the symptoms, the semantic verbal fluency, and the levodopa treatment were the most important for the prediction, and were further used to create a Decision Tree, whose rules may guide the pharmacological management of PD symptoms. Our results contribute to the identification of PD patients who are prone to develop dyskinesia, and may be considered in future clinical trials aiming at developing new therapeutic approaches for PD.
Collapse
Affiliation(s)
- Denisson Augusto Bastos Leal
- Postgraduate Program in Health and Biological Sciences, Federal University of Vale do São Francisco (UNIVASF), Av José Sá de Maniçoba s/n, Petrolina, 56304-917, Brazil
| | - Carla Michele Vieira Dias
- Postgraduate Program in Psychology, Federal University of Vale do São Francisco (UNIVASF), Petrolina, Brazil
| | - Rodrigo Pereira Ramos
- Postgraduate Program in Health and Biological Sciences, Federal University of Vale do São Francisco (UNIVASF), Av José Sá de Maniçoba s/n, Petrolina, 56304-917, Brazil
| | - Ivani Brys
- Postgraduate Program in Health and Biological Sciences, Federal University of Vale do São Francisco (UNIVASF), Av José Sá de Maniçoba s/n, Petrolina, 56304-917, Brazil.
- Postgraduate Program in Psychology, Federal University of Vale do São Francisco (UNIVASF), Petrolina, Brazil.
| |
Collapse
|
16
|
Dias CMV, Leal DAB, Brys I. Levodopa-induced dyskinesia is preceded by increased levels of anxiety and motor impairment in Parkinson's disease patients. Int J Neurosci 2023; 133:1319-1325. [PMID: 35603453 DOI: 10.1080/00207454.2022.2079501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/12/2022] [Indexed: 10/18/2022]
Abstract
Introduction: Dopamine replacement therapy with levodopa is the gold standard treatment of Parkinson's disease (PD); however long-term levodopa use is associated with abnormal involuntary movements known as levodopa-induced dyskinesia (LID) in most patients. LID is not preventable and represents the major limitation of PD treatment.Objective: This study was aimed to find clinical and behavioral features that could be used to identify, years in advance, PD patients that are at high risk of developing LID in the future. Method: Data from PD patients enrolled in The Parkinson's progression markers initiative (PPMI, Michael J. Fox Foundation) that developed dyskinesia during their participation in the study were compared with those who did not, and with healthy controls.Result: LID was preceded byhigher levels of trait anxiety and increased motor impairment in PD patients. Additionally, younger age at PD diagnosis, earlier need for dopaminergic therapy and higher initial levodopa dose, were associated with future development of dyskinesia.Conclusion: These findings suggest that easily detectable clinical and behavioral alterations may help to identify PD patients that are more susceptible to develop LID.
Collapse
Affiliation(s)
- Carla M V Dias
- Research Group in Neuroscience and Experimental Psychology, Petrolina, Pernambuco, Brazil
- Postgraduate Program in Psychology, Federal University of Vale do São Francisco (UNIVASF), Petrolina, Brazil
| | - Denisson A B Leal
- Research Group in Neuroscience and Experimental Psychology, Petrolina, Pernambuco, Brazil
- Postgraduate Program in Health and Biological Sciences, Federal University of Vale do São Francisco (UNIVASF), Petrolina, Brazil
| | - Ivani Brys
- Research Group in Neuroscience and Experimental Psychology, Petrolina, Pernambuco, Brazil
- Postgraduate Program in Psychology, Federal University of Vale do São Francisco (UNIVASF), Petrolina, Brazil
- Postgraduate Program in Health and Biological Sciences, Federal University of Vale do São Francisco (UNIVASF), Petrolina, Brazil
| |
Collapse
|
17
|
Zhai S, Mehrotra DV, Shen J. Applying polygenic risk score methods to pharmacogenomics GWAS: challenges and opportunities. Brief Bioinform 2023; 25:bbad470. [PMID: 38152980 PMCID: PMC10782924 DOI: 10.1093/bib/bbad470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Polygenic risk scores (PRSs) have emerged as promising tools for the prediction of human diseases and complex traits in disease genome-wide association studies (GWAS). Applying PRSs to pharmacogenomics (PGx) studies has begun to show great potential for improving patient stratification and drug response prediction. However, there are unique challenges that arise when applying PRSs to PGx GWAS beyond those typically encountered in disease GWAS (e.g. Eurocentric or trans-ethnic bias). These challenges include: (i) the lack of knowledge about whether PGx or disease GWAS/variants should be used in the base cohort (BC); (ii) the small sample sizes in PGx GWAS with corresponding low power and (iii) the more complex PRS statistical modeling required for handling both prognostic and predictive effects simultaneously. To gain insights in this landscape about the general trends, challenges and possible solutions, we first conduct a systematic review of both PRS applications and PRS method development in PGx GWAS. To further address the challenges, we propose (i) a novel PRS application strategy by leveraging both PGx and disease GWAS summary statistics in the BC for PRS construction and (ii) a new Bayesian method (PRS-PGx-Bayesx) to reduce Eurocentric or cross-population PRS prediction bias. Extensive simulations are conducted to demonstrate their advantages over existing PRS methods applied in PGx GWAS. Our systematic review and methodology research work not only highlights current gaps and key considerations while applying PRS methods to PGx GWAS, but also provides possible solutions for better PGx PRS applications and future research.
Collapse
Affiliation(s)
- Song Zhai
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., North Wales, PA 19454, USA
| | - Judong Shen
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ 07065, USA
| |
Collapse
|
18
|
Rusch C, Flanagan R, Suh H, Subramanian I. To restrict or not to restrict? Practical considerations for optimizing dietary protein interactions on levodopa absorption in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:98. [PMID: 37355689 PMCID: PMC10290638 DOI: 10.1038/s41531-023-00541-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 06/05/2023] [Indexed: 06/26/2023] Open
Abstract
Administration of levodopa for Parkinson's disease (PD) has remained the most effective therapy for symptom management despite being in use for over 50 years. Advancing disease and age, changing tolerability and gastrointestinal (GI) dysfunction may result in change in dietary habits and body weight, as well as unpredictable motor fluctuations and dyskinesias. Dietary proteins which convert into amino acids after digestion are implicated as major factors that inhibit levodopa absorption. For people living with PD (PwP) who experience motor fluctuations, low protein diets (LPD) and protein redistribution diets (PRD) may be effective and are often recommended as a non-pharmacologic approach for improving levodopa bioavailability. However, there is a lack of consensus on a standard definition of these diets and appropriate treatment algorithms for usage. This may be due to the paucity of high-level evidence of LPD and PRD in PwP and whether all or specific subgroups of patients would benefit from these strategies. Managing diet and protein intake with proper education and monitoring may reduce complications associated with these diets such as dyskinesias and unintentional weight loss. Additionally, alterations to medications and GI function may alter levodopa pharmacokinetics. In this narrative review we focus on 1) mechanisms of dietary protein and levodopa absorption in the intestine and blood brain barrier, 2) dietetic approaches to manage protein and levodopa interactions and 3) practical issues for treating PwP as well as future directions to be considered.
Collapse
Affiliation(s)
- C Rusch
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, University of Florida, Gainesville, FL, USA.
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| | | | - H Suh
- Parkinson's Disease Research, Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA, USA
| | - I Subramanian
- Parkinson's Disease Research, Education, and Clinical Center, Greater Los Angeles Veterans Affairs Medical Center, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
19
|
Yan JH, Ge YL, Wang PZ, Li W, Jin H, Zhang JR, Chen J, Wang F, Li D, Mao CJ, Li K, Liu CF. Associations between variants in levodopa metabolic pathway genes and levodopa-induced dyskinesia in Parkinson's disease. Neurosci Lett 2023; 801:137140. [PMID: 36813078 DOI: 10.1016/j.neulet.2023.137140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023]
Abstract
INTRODUCTION Levodopa-induced dyskinesia (LID) is a common motor complication in Parkinson's disease (PD). Several genes in the levodopa metabolic pathway, such as COMT, DRDx and MAO-B, were reported associated with LID. However, there has been no systematic analyses between common variants in levodopa metabolic pathway genes and LID in a large sample of the Chinese population. METHODS Through the whole exome sequencing (WES) and target region sequencing, we aimed to explore the potential associations between common single nucleotide polymorphisms (SNPs) in the levodopa metabolic pathway and LID in Chinese PD individuals. Five hundred and two PD individuals were enrolled in our study, among them, 348 individuals underwent WES, and 154 individuals underwent target region sequencing. We acquired the genetic profile of 11 genes including COMT, DDC, DRD1-5, SLC6A3, TH and MAO-A/B. We established a stepwise strategy to filter SNPs, which finally included 34 SNPs in our analyses. And we used a two-stage study, with discovery (348 individuals with WES) and the replication (all 502 individuals) to confirm our findings. RESULTS Among the 502 PD individuals, 104 (20.7%) were diagnosed with LID. In the discovery stage, we found that COMT rs6269, DRD2 rs6275 and DRD2 rs1076560 were associated with LID. In the replication stage, associations between the three above-mentioned SNPs and LID were still present in all 502 individuals. CONCLUSION We demonstrated that in the Chinese population, COMT rs6269, DRD2 rs6275 and rs1076560 were significantly associated with LID. And rs6275 was reported associated with LID for the first time.
Collapse
Affiliation(s)
- Jia-Hui Yan
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yi-Lun Ge
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Pu-Zhi Wang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wen Li
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong Jin
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin-Ru Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing Chen
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Dan Li
- Department of Neurology, Suqian First People's Hospital, Suqian, Jiangsu, China
| | - Cheng-Jie Mao
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Department of Neurology, Suqian First People's Hospital, Suqian, Jiangsu, China
| | - Kai Li
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Chun-Feng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China; Department of Neurology, Suqian First People's Hospital, Suqian, Jiangsu, China; Department of Neurology, the Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
20
|
Mishima T, Chiu SW, Saiki H, Yamaguchi T, Shimo Y, Maeda T, Watanabe H, Kashihara K, Nomoto M, Hattori N, Tsuboi Y. Risk factors for developing dyskinesia among Parkinson's disease patients with wearing-off: J-FIRST. J Neurol Sci 2023; 448:120619. [PMID: 37023638 DOI: 10.1016/j.jns.2023.120619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/08/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND Dyskinesia frequently occurs during long-term treatment with levodopa in patients with Parkinson's disease (PD) and impacts quality of life. Few studies have examined risk factors for developing dyskinesia in PD patients exhibiting wearing-off. Therefore, we investigated the risk factors and impact of dyskinesia in PD patients exhibiting wearing-off. METHODS We investigated the risk factors and impact of dyskinesia in a 1-year observational study of Japanese PD patients exhibiting wearing-off (J-FIRST). Risk factors were assessed by logistic regression analyses in patients without dyskinesia at study entry. Mixed-effect models were used to evaluate the impact of dyskinesia on changes in Movement Disorder Society-Unified PD Rating Scale (MDS-UPDRS) Part I and PD Questionnaire (PDQ)-8 scores from one timepoint before dyskinesia was observed. RESULTS Of 996 patients analyzed, 450 had dyskinesia at baseline, 133 developed dyskinesia within 1 year, and 413 did not develop dyskinesia. Female sex (odds ratio [95% confidence interval]: 2.636 [1.645-4.223]) and administration of a dopamine agonist (1.840 [1.083-3.126]), a catechol-O-methyltransferase inhibitor (2.044 [1.285-3.250]), or zonisamide (1.869 [1.184-2.950]) were independent risk factors for dyskinesia onset. MDS-UPDRS Part I and PDQ-8 scores increased significantly after the onset of dyskinesia (least-squares mean change [standard error] at 52 weeks: 1.11 [0.52], P = 0.0336; 1.53 [0.48], P = 0.0014; respectively). CONCLUSION Female sex and administration of a dopamine agonist, a catechol-O-methyltransferase inhibitor, or zonisamide were risk factors for dyskinesia onset within 1 year in PD patients exhibiting wearing-off. Nonmotor symptoms and quality of life deteriorated after dyskinesia onset.
Collapse
|
21
|
Chen F, Ren A, Wang M, Fu Y, Huo Y, Chen J, Ge R, Wang H. A case report of Parkinson's disease with acute and unmanageable myoclonic dyskinesia. Clin Neurol Neurosurg 2023; 226:107614. [PMID: 36738642 DOI: 10.1016/j.clineuro.2023.107614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/10/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Affiliation(s)
- Fang Chen
- Department of Neurology, Binzhou Medical University Hospital, China
| | - Anyan Ren
- Department of Neurology, Binzhou Medical University Hospital, China
| | - Mengdi Wang
- Department of Neurology, Binzhou Medical University Hospital, China
| | - Yong Fu
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250000, China
| | - Yingqian Huo
- Department of Neurology, Binzhou Medical University Hospital, China
| | - Jinbo Chen
- Department of Neurology, Binzhou Medical University Hospital, China
| | - Ruli Ge
- Department of Neurology, Binzhou Medical University Hospital, China.
| | - Hongcai Wang
- Department of Neurology, Binzhou Medical University Hospital, China.
| |
Collapse
|
22
|
Dwivedi A, Dwivedi N, Kumar A, Singh VK, Pathak A, Chaurasia RN, Mishra VN, Mohanty S, Joshi D. Association of Catechol-O-Methyltransferase Gene rs4680 Polymorphism and Levodopa Induced Dyskinesia in Parkinson's Disease: A Meta-Analysis and Systematic Review. J Geriatr Psychiatry Neurol 2023; 36:98-106. [PMID: 35603896 DOI: 10.1177/08919887221103580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Long-term levodopa therapy for Parkinson's disease (PD) can cause levodopa induced dyskinesia (LID). Genetic predisposition has a significant role to play in inter-individual heterogeneity in the clinical manifestation of LID. Despite accumulating evidence for the role of COMT gene polymorphism (rs4680) as a genetic basis for LID, to date results have been inconsistent. Early assessment of the Catechol-O-Methyltransferase (COMT) genotype might be helpful to stratify PD patients concerning their individual risk for LID. METHOD In this meta-analysis, we have used 9 studies, which were selected through online databases. Statistical analysis was performed using R (v-3.6) software. 5 genetic models have been used in the present study: Allele model (A vs. G), Dominant model (AA+AG vs. GG), Homozygote model (AA vs. GG), Co-dominant/heterozygote model (AG vs. GG), and Recessive model (AA vs. AG + GG). RESULTS The results indicated a significant association between COMT rs4680 (Val158Met) polymorphism and LID risk. The genotype AA of COMT rs4680 is a risk factor for LID in PD patients under the recessive model (AA vs GG+AG) in the random-effect model. Analysis based on ethnicity showed that COMT rs4680 SNP allele A is a risk factor for LID development in Asian PD patients, while GG genotype is a risk factor for LID development in non-Asian PD patients using different genetic models. CONCLUSION The results of the present meta-analysis support that the COMT Val158Met polymorphism is a risk factor for the development of LID in PD patients having ethnic variations.
Collapse
Affiliation(s)
- Archana Dwivedi
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| | - Nidhi Dwivedi
- Department of community medicine, NDMC Medical College and 56888Hindu Rao Hospital, New Delhi, India
| | - Anand Kumar
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| | - Varun K Singh
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| | - Abhishek Pathak
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| | - R N Chaurasia
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| | - V N Mishra
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, 28730All India Institute of Medical Sciences, New Delhi, India
| | - Deepika Joshi
- Department of Neurology, Institute of Medical Sciences, 30117Banaras Hindu University, Varanasi, India
| |
Collapse
|
23
|
Yan JH, Li K, Ge YL, Li W, Wang PZ, Jin H, Zhang JR, Chen J, Wang F, Yang YP, Zhang YC, Li D, Mao CJ, Liu CF. Quantitative Transcranial Sonography Evaluation of Substantia Nigra Hyperechogenicity Is Useful for Predicting Levodopa-Induced Dyskinesia in Parkinson Disease. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:607-615. [PMID: 36456377 DOI: 10.1016/j.ultrasmedbio.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/07/2022] [Accepted: 10/30/2022] [Indexed: 06/17/2023]
Abstract
Levodopa-induced dyskinesia (LID) is a common motor complication in Parkinson disease (PD). Abnormal substantia nigra hyperechogenicity (SN+), detected by transcranial sonography (TCS), plays an important role in the differential diagnosis of PD. The purpose of this study was to investigate the predictive performance of quantitative SN+ evaluations for LID. Five hundred sixty-two individuals were included in our analysis, and 198 individuals were followed up. These individuals were divided into two groups at baseline: the PD with LID (PD+LID) group and the PD without LID (PD-LID) group. The association between total hyperechogenic area of the SN on both sides (SNT) and LID was analyzed by binary logistic analysis. A binary logistic regression model including SNT was applied to establish a model for discriminating LID. At baseline, 105 (18.7%) individuals were diagnosed with LID. The PD+LID group had a longer disease duration, shorter education duration, higher levodopa equivalent doses, greater disease severity and larger SNT. A model combining clinical features and SNT was further established with better efficiency (area under the receiver operating characteristic curve = 0.839). One hundred ninety-eight individuals were followed up; individuals with a larger SNT and a higher predicted probability were more likely to develop LID in our follow-up. Our study determined that quantitative TCS evaluation of SN echogenicity is useful in predicting LID in PD.
Collapse
Affiliation(s)
- Jia-Hui Yan
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Kai Li
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yi-Lun Ge
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wen Li
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Pu-Zhi Wang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong Jin
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin-Ru Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing Chen
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Ya-Ping Yang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying-Chun Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dan Li
- Department of Neurology, Suqian First People's Hospital, Suqian, Jiangsu, China
| | - Cheng-Jie Mao
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Department of Neurology, Suqian First People's Hospital, Suqian, Jiangsu, China
| | - Chun-Feng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China; Department of Neurology, Suqian First People's Hospital, Suqian, Jiangsu, China; Department of Neurology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
24
|
Hattori N, Kamei T, Ishida T, Suzuki I, Nomoto M, Tsuboi Y. Long-term effects of safinamide adjunct therapy on levodopa-induced dyskinesia in Parkinson's disease: post-hoc analysis of a Japanese phase III study. J Neural Transm (Vienna) 2022; 129:1277-1287. [PMID: 36001147 PMCID: PMC9468087 DOI: 10.1007/s00702-022-02532-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/19/2022] [Indexed: 11/03/2022]
Abstract
This post-hoc analysis investigated the long-term effects of safinamide on the course of dyskinesia and efficacy outcomes using data from a phase III, open-label 52-week study of safinamide 50 or 100 mg/day in Japanese patients with Parkinson’s disease (PD) with wearing-off. Patients (N = 194) were grouped using the UPDRS Part IV item 32: with and without pre-existing dyskinesia (pre-D subgroup; item 32 > 0 at baseline [n = 81], without pre-D subgroup; item 32 = 0 at baseline [n = 113]). ON-time with troublesome dyskinesia (ON-TD) increased significantly from baseline to Week 4 in the pre-D subgroup (+ 0.25 ± 0.11 h [mean ± SE], p = 0.0355) but gradually decreased up to Week 52 (change from baseline: − 0.08 ± 0.17 h, p = 0.6224); ON-TD did not change significantly in the Without pre-D subgroup. UPDRS Part IV item 32 score increased significantly at Week 52 compared with baseline in the Without pre-D subgroup, but no UPDRS Part IV dyskinesia related-domains changed in the pre-D subgroup. Both subgroups improved in ON-time without TD, UPDRS Part III, and Part II [OFF-phase] scores. The cumulative incidence of new or worsening dyskinesia (adverse drug reaction) at Week 52 was 32.5 and 5.0% in the pre-D and Without pre-D subgroups, respectively. This study suggested that safinamide led to short-term increasing dyskinesia but may be not associated with marked dyskinesia at 1-year follow-up in patients with pre-existing dyskinesia, and that it improved motor symptoms regardless of the presence or absence of dyskinesia at baseline. Further studies are warranted to investigate this association in more details. Trial registration: JapicCTI-153057 (Registered: 2015/11/02).
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.
| | - Takanori Kamei
- Medical Headquarters, Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo, 112-8088, Japan
| | - Takayuki Ishida
- Medical Headquarters, Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo, 112-8088, Japan
| | - Ippei Suzuki
- Medicine Development, Deep Human Biology Learning, Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo, 112-8088, Japan
| | - Masahiro Nomoto
- Saiseikai Imabari Center for Health and Welfare, 7-6-1 Kitamura, Imabari, Ehime, 799-1592, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
25
|
Zhang H, Wang L, Gan C, Cao X, Ji M, Sun H, Yuan Y, Zhang K. Altered functional connectivity of cerebellar dentate nucleus in peak-dose dyskinesia in Parkinson’s disease. Front Aging Neurosci 2022; 14:943179. [PMID: 36034152 PMCID: PMC9400811 DOI: 10.3389/fnagi.2022.943179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The cerebellum is associated with the emergence of levodopa-induced dyskinesia (LID) in Parkinson’s disease (PD), yet the neural mechanism remains obscure. Our aim was to ascertain the role of functional connectivity (FC) patterns of the cerebellar dentate nucleus (DN) in the pathogenesis of peak-dose dyskinesia in PD. Twenty-three peak-dose dyskinetic PD patients, 27 non-dyskinetic PD patients, and 36 healthy controls (HCs) were enrolled and underwent T1-weighted and resting-state functional magnetic resonance imaging (rs-fMRI) scans after dopaminergic medication intake. We selected left and right DN as the regions of interest and then employed voxel-wise FC analysis and voxel-based morphometry analysis (VBM). The correlations between the altered FC pattern and clinical scores were also examined. Finally, receiver operating characteristic (ROC) curve analysis was performed to assess the potential of DN FC measures as a feature of peak-dose dyskinesia in PD. Dyskinetic PD patients showed excessively increased FC between the left DN and right putamen compared with the non-dyskinetic. When compared with controls, dyskinetic PD patients mainly exhibited increased FC between left DN and bilateral putamen, left paracentral lobule, right postcentral gyrus, and supplementary motor area. Additionally, non-dyskinetic PD patients displayed increased FC between left DN and left precentral gyrus and right paracentral lobule compared with controls. Meanwhile, increased FC between DN (left/right) and ipsilateral cerebellum lobule VIII was observed in both PD subgroups. However, no corresponding alteration in gray matter volume (GMV) was found. Further, a positive correlation between the z-FC values of left DN-right putamen and the Unified Dyskinesia Rating Scale (UDysRS) was confirmed in dyskinetic PD patients. Notably, ROC curve analyses revealed that the z-FC values of left DN-right putamen could be a potential neuroimaging feature identifying dyskinetic PD patients. Our findings demonstrated that the excessively strengthened connectivity of DN-putamen might contribute to the pathophysiological mechanisms of peak-dose dyskinesia in PD.
Collapse
|
26
|
Hill EJ, Robak LA, Al-Ouran R, Deger J, Fong JC, Vandeventer PJ, Schulman E, Rao S, Saade H, Savitt JM, von Coelln R, Desai N, Doddapaneni H, Salvi S, Dugan-Perez S, Muzny DM, McGuire AL, Liu Z, Gibbs RA, Shaw C, Jankovic J, Shulman LM, Shulman JM. Genome Sequencing in the Parkinson Disease Clinic. Neurol Genet 2022; 8:e200002. [PMID: 35747619 PMCID: PMC9210549 DOI: 10.1212/nxg.0000000000200002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
Abstract
Background and Objectives Genetic variants affect both Parkinson disease (PD) risk and manifestations. Although genetic information is of potential interest to patients and clinicians, genetic testing is rarely performed during routine PD clinical care. The goal of this study was to examine interest in comprehensive genetic testing among patients with PD and document reactions to possible findings from genome sequencing in 2 academic movement disorder clinics. Methods In 203 subjects with PD (age = 63 years, 67% male), genome sequencing was performed and filtered using a custom panel, including 49 genes associated with PD, parkinsonism, or related disorders, as well as a 90-variant PD genetic risk score. Based on the results, 231 patients (age = 67 years, 63% male) were surveyed on interest in genetic testing and responses to vignettes covering (1) familial risk of PD (LRRK2); (2) risk of PD dementia (GBA); (3) PD genetic risk score; and (4) secondary, medically actionable variants (BRCA1). Results Genome sequencing revealed a LRRK2 variant in 3% and a GBA risk variant in 10% of our clinical sample. The genetic risk score was normally distributed, identifying 41 subjects with a high risk of PD. Medically actionable findings were discovered in 2 subjects (1%). In our survey, the majority (82%) responded that they would share a LRRK2 variant with relatives. Most registered unchanged or increased interest in testing when confronted with a potential risk for dementia or medically actionable findings, and most (75%) expressed interest in learning their PD genetic risk score. Discussion Our results highlight broad interest in comprehensive genetic testing among patients with PD and may facilitate integration of genome sequencing in clinical practice.
Collapse
Affiliation(s)
| | | | - Rami Al-Ouran
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Jennifer Deger
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Jamie C. Fong
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Paul Jerrod Vandeventer
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Emily Schulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Sindhu Rao
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Hiba Saade
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joseph M. Savitt
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Rainer von Coelln
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Neeja Desai
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Harshavardhan Doddapaneni
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Sejal Salvi
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Shannon Dugan-Perez
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Donna M. Muzny
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Amy L. McGuire
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Zhandong Liu
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Richard A. Gibbs
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Chad Shaw
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joseph Jankovic
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Lisa M. Shulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joshua M. Shulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| |
Collapse
|
27
|
Jeong SH, Lee HS, Lee PH, Sohn YH, Chung SJ. Does dopamine deficiency affect sex-dependent prognosis in Parkinson's disease? Parkinsonism Relat Disord 2022; 102:57-63. [DOI: 10.1016/j.parkreldis.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/11/2022] [Accepted: 07/17/2022] [Indexed: 10/16/2022]
|
28
|
Jung JH, Kim YJ, Chung SJ, Yoo HS, Lee YH, Baik K, Jeong SH, Lee YG, Lee HS, Ye BS, Sohn YH, Jeong Y, Lee PH. White matter connectivity networks predict levodopa-induced dyskinesia in Parkinson's disease. J Neurol 2022; 269:2948-2960. [PMID: 34762146 DOI: 10.1007/s00415-021-10883-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Although levodopa-induced dyskinesia-relevant white matter change has been evaluated, it is uncertain whether these changes may reflect the underlying predisposing conditions leading to the development of levodopa-induced dyskinesia. OBJECTIVE To elucidate the role of white matter connectivity networks in the development of levodopa-induced dyskinesia in drug-naïve Parkinson's disease. METHODS We recruited 30 patients who developed levodopa-induced dyskinesia within 5 years from MRI acquisition (vulnerable-group), 47 patients who had not developed levodopa-induced dyskinesia within 5 years (resistant-group), and 28 controls. We performed comparative analyses of whole-brain white matter integrity and connectivity using tract-based spatial and network- and degree-based statistics. We evaluated the predictability of levodopa-induced dyskinesia development and relationship with its latency, using the average connectivity strength as a predictor in Cox- and linear-regression, respectively. RESULTS Mean-diffusivity was lower mainly at the left frontal region in the vulnerable-group compared to the resistant-group. Network-based statistics identified a subnetwork consisting of the bilateral fronto-striato-pallido-thalamic and lateral parietal regions (subnetwork A) and degree-based statistics identified four subnetworks (hub-subnetwork) consisting of edges centered on the left superior frontal gyrus, left putamen, left insular, or left precentral gyrus, where the vulnerable-group had stronger connectivity compared to the resistant-group. Stronger connectivity within the subnetwork A and hub-subnetwork centered on the left superior frontal gyrus was a predictor of levodopa-induced dyskinesia development independent of known risk factors and had an inverse relationship with its latency. CONCLUSIONS Our data suggest that white matter connectivity subnetworks within corticostriatal regions play a pivotal role in the development of levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Jin Ho Jung
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Yae Ji Kim
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Han Soo Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yang Hyun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoungwon Baik
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seong Ho Jeong
- Department of Neurology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Young Gun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong Jeong
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
29
|
Baba Y, Futamura A, Kinno R, Nomoto S, Takahashi S, Yasumoto T, Osakabe Y, Shoji D, Nabeshima Y. The relationship between the distinct ratios of benserazide and carbidopa to levodopa and motor complications in Parkinson's disease: A retrospective cohort study. J Neurol Sci 2022; 437:120263. [DOI: 10.1016/j.jns.2022.120263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/16/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
|
30
|
Bove F, Calabresi P. Plasticity, genetics, and epigenetics in l-dopa-induced dyskinesias. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:167-184. [PMID: 35034732 DOI: 10.1016/b978-0-12-819410-2.00009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
l-Dopa-induced dyskinesias (LIDs) are a frequent complication in l-dopa-treated patients affected by Parkinson's disease (PD). In the last years, several progresses in the knowledge of LIDs mechanisms have led to the identification of several molecular and electrophysiologic events. A complex cascade of intracellular events underlies the pathophysiology of LIDs, and, among these, aberrant plasticity in the cortico-basal ganglia system, at striatal and cortical level, plays a key role. Furthermore, several recent studies have investigated genetic susceptibility and epigenetic modifications in LIDs pathophysiology that might have future relevance in clinical practice and pharmacologic research. These progresses might lead to the development of specific strategies not only to treat, but also to prevent or delay the development of LIDs in PD.
Collapse
Affiliation(s)
- Francesco Bove
- UOC Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paolo Calabresi
- UOC Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
31
|
Won JH, Youn J, Park H. Enhanced neuroimaging genetics using multi-view non-negative matrix factorization with sparsity and prior knowledge. Med Image Anal 2022; 77:102378. [DOI: 10.1016/j.media.2022.102378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/29/2021] [Accepted: 01/26/2022] [Indexed: 11/28/2022]
|
32
|
Johnson D, Wilke MA, Lyle SM, Kowalec K, Jorgensen A, Wright GE, Drögemöller BI. A systematic review and analysis of the use of polygenic scores in pharmacogenomics. Clin Pharmacol Ther 2021; 111:919-930. [PMID: 34953075 DOI: 10.1002/cpt.2520] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/18/2021] [Indexed: 11/09/2022]
Abstract
Polygenic scores (PGS) have emerged as promising tools for complex trait risk prediction. The application of these scores to pharmacogenomics provides new opportunities to improve the prediction of treatment outcomes. To gain insight into this area of research, we conducted a systematic review and accompanying analysis. This review uncovered 51 papers examining the use of PGS for drug-related outcomes, with the majority of these papers focusing on the treatment of psychiatric disorders (n=30). Due to difficulties in collecting large cohorts of uniformly treated patients, the majority of pharmacogenomic PGS were derived from large-scale genome-wide association studies of disease phenotypes that were related to the pharmacogenomic phenotypes under investigation (e.g. schizophrenia-derived PGS for antipsychotic response prediction). Examination of the research participants included in these studies revealed that the majority of cohort participants were of European descent (78.4%). These biases were also reflected in research affiliations, which were heavily weighted towards institutions located in Europe and North America, with no first or last authors originating from institutions in Africa or South Asia. There was also substantial variability in the methods used to develop PGS, with between 3 and 6.6 million variants included in the PGS. Finally, we observed significant inconsistencies in the reporting of PGS analyses and results, particularly in terms of risk model development and application, coupled with a lack of data transparency and availability, with only three pharmacogenomics PGS deposited on the PGS Catalog. These findings highlight current gaps and key areas for future pharmacogenomic PGS research.
Collapse
Affiliation(s)
- Danielle Johnson
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - MacKenzie Ap Wilke
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Sarah M Lyle
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Kaarina Kowalec
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Jorgensen
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Galen Eb Wright
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre and Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,CancerCare Manitoba Research Institute, Winnipeg, MB, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
33
|
Zhang Z, Liu G, Wang D, Chen H, Su D, Kou W, Zhao J, Wang X, Wang Z, Ma H, Feng T. Effect of onset age on the levodopa threshold dosage for dyskinesia in Parkinson's disease. Neurol Sci 2021; 43:3165-3174. [PMID: 34853899 DOI: 10.1007/s10072-021-05694-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION With the levodopa threshold effect for dyskinesia observed, threshold dosage of levodopa was identified in the general Parkinson's disease (PD) population. While early-onset PD (EOPD) and late-onset PD (LOPD) differ in the pathogenesis and clinical manifestations, threshold dosage of levodopa for individualized treatment remains unestablished. The objective of this study was to propose threshold dosage of levodopa in EOPD and LOPD patients, respectively. METHODS Data on demographic and clinical and treatment measures were collected in 539 PD patients. Patients were divided into different onset groups using 50 as the cut-off age. We used univariable and multivariable analysis to screen for risk factors for dyskinesia. Receiver operating characteristic curve was used to determine the levodopa threshold dosages for dyskinesia. RESULTS The prevalence of dyskinesia was 47.7% (53/111) in the EOPD group and 24.1% (103/428) in the LOPD group. Risk factors identified for dyskinesia include high levodopa daily dose and levodopa responsiveness for EOPD patients and high levodopa daily dose, long levodopa treatment duration, low body weight, use of entacapone, and high Hoehn-Yahr stage in off state for LOPD patients. The daily levodopa threshold dosages were 400 mg or 5.9 mg/kg for EOPD and 450 mg or 7.2 mg/kg for LOPD. CONCLUSION EOPD patients had lower levodopa threshold dosage comparing with LOPD patients. Treatment of EOPD requires stricter levodopa dose control to delay the onset of dyskinesia.
Collapse
Affiliation(s)
- Zhijin Zhang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Genliang Liu
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dongxu Wang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huimin Chen
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dongning Su
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wenyi Kou
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiajia Zhao
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xuemei Wang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhan Wang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huizi Ma
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China. .,China National Clinical Research Center for Neurological Diseases, Beijing, China. .,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
34
|
Iyer V, Venkiteswaran K, Savaliya S, Lieu CA, Handly E, Gilmour TP, Kunselman AR, Subramanian T. The cross-hemispheric nigrostriatal pathway prevents the expression of levodopa-induced dyskinesias. Neurobiol Dis 2021; 159:105491. [PMID: 34461264 PMCID: PMC8597404 DOI: 10.1016/j.nbd.2021.105491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/17/2021] [Accepted: 08/25/2021] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative movement disorder that is routinely treated with levodopa. Unfortunately, long-term dopamine replacement therapy using levodopa leads to levodopa-induced dyskinesias (LID), a significant and disabling side-effect. Clinical findings indicate that LID typically only occurs following the progression of PD motor symptoms from the unilateral (Hoehn and Yahr (HY) Stage I) to the bilateral stage (HY Stage II). This suggests the presence of some compensatory interhemispheric mechanisms that delay the occurrence of LID. We therefore investigated the role of interhemispheric connections of the nigrostriatal pathway on LID expression in a rat model of PD. The striatum of one hemisphere of rats was first injected with a retrograde tracer to label the ipsi- and cross-hemispheric nigrostriatal pathways. Rats were then split into groups and unilaterally lesioned in the striatum or medial forebrain bundle of the tracer-injected hemisphere to induce varying levels of hemiparkinsonism. Finally, rats were treated with levodopa and tested for the expression of LID. Distinct subsets emerged from rats that underwent the same lesioning paradigm based on LID. Strikingly, non-dyskinetic rats had significant sparing of their cross-hemispheric nigrostriatal pathway projecting from the unlesioned hemisphere. In contrast, dyskinetic rats only had a small proportion of this cross-hemispheric nigrostriatal pathway survive lesioning. Crucially, both non-dyskinetic and dyskinetic rats had nearly identical levels of ipsi-hemispheric nigrostriatal pathway survival and parkinsonian motor deficits. Our data suggest that the survival of the cross-hemispheric nigrostriatal pathway plays a crucial role in preventing the expression of LID and represents a potentially novel target to halt the progression of this devastating side-effect of a common anti-PD therapeutic.
Collapse
Affiliation(s)
- Vishakh Iyer
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Kala Venkiteswaran
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Sandip Savaliya
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Christopher A Lieu
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Erin Handly
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Timothy P Gilmour
- Department of Electrical Engineering, John Brown University, Siloam Springs, AR, United States of America
| | - Allen R Kunselman
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Thyagarajan Subramanian
- Department of Neurology and Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, United States of America.
| |
Collapse
|
35
|
Levodopa-Induced Ocular Dyskinesia in an Early-Onset Parkinson Disease Patient With GBA Mutation. Clin Neuropharmacol 2021; 44:201-204. [PMID: 34654015 PMCID: PMC8594500 DOI: 10.1097/wnf.0000000000000484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental digital content is available in the text. Objectives The aim of this study was to report a case of levodopa-induced ocular dyskinesia in an early-onset Parkinson disease patient and to investigate the pathogenic gene. Methods We report the case of a 49-year-old male patient with a 13-year history of Parkinson disease. Involuntary eye movements were noticed after treatment with amantadine for limb dyskinesias. Levodopa-induced ocular dyskinesias involving repetitive, transient, and stereotyped rightward deviations of gaze appeared after intake of an antiparkinsonian drug. Limb dyskinesias also occurred simultaneously. We used a next-generation sequencing targeted gene panel and found a heterozygous missense mutation (p.R535H) in GBA. Direct Sanger sequencing verified the missense mutation. Conclusions We report the case of an uncommon early-onset PD patient carrying a GBA mutation presenting ocular dyskinesia. Genetic screening may provide a better mechanistic insight into dyskinesias.
Collapse
|
36
|
Polygenic Risk Scores Contribute to Personalized Medicine of Parkinson's Disease. J Pers Med 2021; 11:jpm11101030. [PMID: 34683174 PMCID: PMC8539098 DOI: 10.3390/jpm11101030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/18/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder characterized by the loss of dopaminergic neurons. The vast majority of PD patients develop the disease sporadically and it is assumed that the cause lies in polygenic and environmental components. The overall polygenic risk is the result of a large number of common low-risk variants discovered by large genome-wide association studies (GWAS). Polygenic risk scores (PRS), generated by compiling genome-wide significant variants, are a useful prognostic tool that quantifies the cumulative effect of genetic risk in a patient and in this way helps to identify high-risk patients. Although there are limitations to the construction and application of PRS, such as considerations of limited genetic underpinning of diseases explained by SNPs and generalizability of PRS to other populations, this personalized risk prediction could make a promising contribution to stratified medicine and tailored therapeutic interventions in the future.
Collapse
|
37
|
Hutny M, Hofman J, Klimkowicz-Mrowiec A, Gorzkowska A. Current Knowledge on the Background, Pathophysiology and Treatment of Levodopa-Induced Dyskinesia-Literature Review. J Clin Med 2021; 10:jcm10194377. [PMID: 34640395 PMCID: PMC8509231 DOI: 10.3390/jcm10194377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/02/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
Levodopa remains the primary drug for controlling motor symptoms in Parkinson’s disease through the whole course, but over time, complications develop in the form of dyskinesias, which gradually become more frequent and severe. These abnormal, involuntary, hyperkinetic movements are mainly characteristic of the ON phase and are triggered by excess exogenous levodopa. They may also occur during the OFF phase, or in both phases. Over the past 10 years, the issue of levodopa-induced dyskinesia has been the subject of research into both the substrate of this pathology and potential remedial strategies. The purpose of the present study was to review the results of recent research on the background and treatment of dyskinesia. To this end, databases were reviewed using a search strategy that included both relevant keywords related to the topic and appropriate filters to limit results to English language literature published since 2010. Based on the selected papers, the current state of knowledge on the morphological, functional, genetic and clinical features of levodopa-induced dyskinesia, as well as pharmacological, genetic treatment and other therapies such as deep brain stimulation, are described.
Collapse
Affiliation(s)
- Michał Hutny
- Students’ Scientific Society, Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
- Correspondence:
| | - Jagoda Hofman
- Students’ Scientific Society, Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Aleksandra Klimkowicz-Mrowiec
- Department of Internal Medicine and Gerontology, Faculty of Medicine, Medical College, Jagiellonian University, 30-688 Kraków, Poland;
| | - Agnieszka Gorzkowska
- Department of Neurorehabilitation, Faculty of Medical Sciences, School of Medicine, Medical University of Silesia, 40-752 Katowice, Poland;
| |
Collapse
|
38
|
Simoni S, Paoletti FP, Eusebi P, Cappelletti G, Filidei M, Brahimi E, Nigro P, Santangelo V, Parnetti L, Calabresi P, Tambasco N. Impulse Control Disorders and Levodopa-Induced Dyskinesias in Parkinson's Disease: Pulsatile versus Continuous Dopaminergic Stimulation. JOURNAL OF PARKINSONS DISEASE 2021; 10:927-934. [PMID: 32280105 DOI: 10.3233/jpd-191833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dopaminergic medications in Parkinson's disease (PD) are usually associated with the development of both levodopa-induced dyskinesias (LID) and impulse control and repetitive behavior disorders (ICRB). OBJECTIVE To assess the prevalence and the severity of ICRB in a cohort of moderate and advanced PD patients and to investigate the potential interplay between ICRB, LID and dopaminergic therapies. METHODS 117 PD patients were consecutively recruited. LID were assessed by using the Rush Dyskinesia Rating Scale (RDRS). ICRB were tested by means of Questionnaire for Impulsive Compulsive Disorders in Parkinson's Disease Rating Scale (QUIP-RS). RESULTS 55 patients were affected by LID. Among them, 37 were treated only by oral therapy, OT (LID/OT), while 18 were on treatment with jejunal levodopa infusion, JLI (LID/JLI). 62 patients were not affected by LID (NLID) and all of them were on therapy only with oral drugs. The overall prevalence of clinically significant ICRB was 34% (95% CI = 26% to 43%) and the mean value (±SD) of QUIP-RS total score was 5.4±8.5. Prevalence of clinically significant ICRB, as well as severity of ICRB, was higher in patients with LID compared to NLID patients (p = 0.016 and p < 0.001, respectively). When considering LID/JLI, LID/OT and NLID groups, QUIP-RS total score was significantly higher in LID/OT patients compared to LID/JLI (10.4±11.8 vs. 4.9±6.0, p = 0.019) and NLID (10.4±11.8 vs. 2.5±4.8, p < 0.001) groups. CONCLUSION PD patients with LID show ICRB more frequently and more severely than patients without LID. Among LID patients, those treated by JLI showed a lower severity of ICRB than those on OT, suggesting a potential protective effect of JLI on ICRB.
Collapse
Affiliation(s)
- Simone Simoni
- Movement Disorders Center, Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Federico Paolini Paoletti
- Movement Disorders Center, Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy.,Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Paolo Eusebi
- Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Giulia Cappelletti
- Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Marta Filidei
- Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Elona Brahimi
- Neurology Department, Regina Montis Regalis Hospital, Cuneo, Italy
| | - Pasquale Nigro
- Movement Disorders Center, Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Valerio Santangelo
- Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy.,Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Lucilla Parnetti
- Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy.,Section of Neurology Department, Center for Memory Disturbances, Laboratory of Clinical Neurochemistry, Perugia General Hospital and University of Perugia, Perugia, Italy
| | - Paolo Calabresi
- Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nicola Tambasco
- Movement Disorders Center, Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy.,Neurology Department, Perugia General Hospital and University of Perugia, Perugia, Italy
| |
Collapse
|
39
|
Association of COMT rs4680 and MAO-B rs1799836 polymorphisms with levodopa-induced dyskinesia in Parkinson's disease-a meta-analysis. Neurol Sci 2021; 42:4085-4094. [PMID: 34346015 DOI: 10.1007/s10072-021-05509-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/18/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND PURPOSE Polymorphisms of the catechol-O-methyl transferase (COMT) or monoamine oxidase B (MAO-B) genes may affect the occurrence of dyskinesia in Parkinson's disease (PD) patients. However, the findings are inconsistent. Thus, we performed a meta-analysis to assess whether COMT and MAO-B genetic variants are associated with an increased incidence of levodopa-induced dyskinesia (LID) in PD patients. METHODS A literature search of PubMed, Embase, and Cochrane Library was conducted to identify relevant studies published up to January 2021. The strength of the association between the polymorphisms and LID susceptibility was estimated by odds ratio (OR) and associated 95% confidence interval (CI). The pooled ORs were assessed in different genetic models. RESULTS Ten studies involving 2385 PD patients were included in the meta-analysis. Analysis of pooled ORs and 95% CIs suggested that the AA genotype of COMT(rs4680) was associated with LID (OR = 1.39, 95%CI: 1.02-1.89, P = 0.039) in the recessive model, and this correlation was more obvious in Brazilian samples in the analysis stratified by ethnicity. For the AG genotype of MAO-B(rs1799836), the pooled OR was 1.66 (95% CI: 1.04-2.65, P = 0.03) in patients with LID versus those without LID in the heterozygote model. CONCLUSIONS Our meta-analysis implicates the AA genotype of the COMT rs4680 polymorphism as potentially increasing the risk of LID in a recessive genetic model for PD patients. Furthermore, the AG genotype of the MAO-B rs1799836 polymorphism may influence the prevalence of LID in PD patients in the heterozygote model. However, further well-designed studies with larger PD patient cohorts are required to validate these results after adjusting for confounding factors.
Collapse
|
40
|
Cui J, Qin Y, Tian Y, Ge X, Han H, Yang Z, Yu H. Activities of daily living as a longitudinal moderator of the effect of autonomic dysfunction on anxiety and depression of Parkinson's patients. Brain Behav 2021; 11:e2297. [PMID: 34333879 PMCID: PMC8413789 DOI: 10.1002/brb3.2297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND There is no clear time point for the onset of depression and anxiety in Parkinson's disease (PD), and their atypical physical symptoms often overlap with other nonmotor symptoms. Autonomic dysfunction usually appears earlier than motor symptoms, seriously impairing activities of daily living (ADL), even quality of life. Whether autonomic dysfunction can affect depression and anxiety in PD patients through ADL is still unclear. METHODS We conducted three progressive autoregressive mediation models to evaluate whether ADL may mediate the association between autonomic symptom burden, where the mediation chain with autonomic function as an independent variable, ADL as a mediating variable, and anxiety and depression as dependent variables. The ADL of PD patients were measured by the Scales for Outcomes in Parkinson's disease-Autonomic (SCOPA-AUT) and Modified Schwab and England ADL scale, respectively, and the status of depression and anxiety were measured by the Geriatric Depression Scale (GDS) and State-Trait Anxiety Inventory (STAI). RESULTS There were 338 PD patients, including 220 males and 118 females. Demographic information, including age, gender, and education level, were not correlated with the depression and anxiety. Model III had the smallest AIC (AIC = 12,669.89), and the cross-lagged relations were not statistically significant, so we selected Model II as the optimal model. In Model II, longitudinal autoregressive mediated effect and longitudinal mediated effect of autonomic dysfunction affecting anxiety and depression through ADL were not statistically significant, suggesting longitudinal changes of autonomic dysfunction were independent of anxiety and depression through ADL. Contemporaneous mediated effects of autonomic dysfunction affecting anxiety and depression through ADL were statistically significant, suggesting contemporaneous autonomic dysfunction may contribute to anxiety and depression through ADL. CONCLUSIONS Targeted prevention and intervention measures for autonomic dysfunction and ADL should be taken to preserve and improve self-perceived life satisfaction in the clinical practice and preventive health care of PD.
Collapse
Affiliation(s)
- Jing Cui
- Department of Health StatisticsSchool of Public HealthShanxi Medical UniversityTaiyuanChina
| | - Yao Qin
- Department of Health StatisticsSchool of Public HealthShanxi Medical UniversityTaiyuanChina
| | - Yuling Tian
- Department of NeurologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Xiaoyan Ge
- Department of Health StatisticsSchool of Public HealthShanxi Medical UniversityTaiyuanChina
| | - Hongjuan Han
- Department of Health StatisticsSchool of Public HealthShanxi Medical UniversityTaiyuanChina
| | - Zongfang Yang
- Department of Health StatisticsSchool of Public HealthShanxi Medical UniversityTaiyuanChina
| | - Hongmei Yu
- Department of Health StatisticsSchool of Public HealthShanxi Medical UniversityTaiyuanChina
- Shanxi Provincial Key Laboratory of Major Diseases Risk AssessmentTaiyuanChina
| |
Collapse
|
41
|
Chen YP, Ou RW, Gu XJ, Zhang LY, Cao B, Hou YB, Liu KC, Lin JY, Wei QQ, Zhao B, Wu Y, Shang HF. Multivariable clinical-genetic model for predicting dyskinesia in early-onset Parkinson's disease. Transl Neurodegener 2021; 10:26. [PMID: 34325746 PMCID: PMC8320054 DOI: 10.1186/s40035-021-00251-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/15/2021] [Indexed: 02/08/2023] Open
Affiliation(s)
- Yong-Ping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ru-Wei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao-Jing Gu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ling-Yu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan-Bing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kun-Cheng Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jun-Yu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qian-Qian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Wu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui-Fang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
42
|
Studying the functional connectivity of the primary motor cortex with the binarized cross recurrence plot: The influence of Parkinson's disease. PLoS One 2021; 16:e0252565. [PMID: 34097691 PMCID: PMC8183987 DOI: 10.1371/journal.pone.0252565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/18/2021] [Indexed: 11/20/2022] Open
Abstract
Two new recurrence plot methods (the binary recurrence plot and binary cross recurrence plot) were introduced here to study the long-term dynamic of the primary motor cortex and its interaction with the primary somatosensory cortex, the anterior motor thalamus of the basal ganglia motor loop and the precuneous nucleus of the default mode network. These recurrence plot methods: 1. identify short-term transient interactions; 2. identify long-lasting delayed interactions that are common in complex systems; 3. work with non-stationary blood oxygen level dependent (BOLD) data; 4. may study the relationship of centers with non-linear functional interactions; 5 may compare different experimental groups performing different tasks. These methods were applied to BOLD time-series obtained in 20 control subjects and 20 Parkinson´s patients during the execution of motor activity and body posture tasks (task-block design). The binary recurrence plot showed the task-block BOLD response normally observed in the primary motor cortex with functional magnetic resonance imaging methods, but also shorter and longer BOLD-fluctuations than the task-block and which provided information about the long-term dynamic of this center. The binary cross recurrence plot showed short-lasting and long-lasting functional interactions between the primary motor cortex and the primary somatosensory cortex, anterior motor thalamus and precuneous nucleus, interactions which changed with the resting and motor tasks. Most of the interactions found in healthy controls were disrupted in Parkinson's patients, and may be at the basis of some of the motor disorders and side-effects of dopaminergic drugs commonly observed in these patients.
Collapse
|
43
|
Tirozzi A, Modugno N, Palomba NP, Ferese R, Lombardi A, Olivola E, Gialluisi A, Esposito T. Analysis of Genetic and Non-genetic Predictors of Levodopa Induced Dyskinesia in Parkinson's Disease. Front Pharmacol 2021; 12:640603. [PMID: 33995045 PMCID: PMC8118664 DOI: 10.3389/fphar.2021.640603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/14/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Levodopa (L-Dopa), representing the therapeutic gold standard for the treatment of Parkinson disease (PD), is associated with side effects like L-Dopa induced dyskinesia (LID). Although several non-genetic and genetic factors have been investigated for association with LID risk, contrasting results were reported and its genetic basis remain largely unexplored. Methods: In an Italian PD cohort (N = 460), we first performed stepwise multivariable Cox Proportional Hazard regressions modeling LID risk as a function of gender, PD familiarity, clinical subtype, weight, age-at-onset (AAO) and years-of-disease (YOD), L-Dopa dosage, severity scores, and scales assessing motor (UPDRS-III), cognitive (MoCA), and non-motor symptoms (NMS). Then we enriched the resulting model testing two variants—rs356219 and D4S3481—increasing the expression of the SNCA gene, previously suggested as a potential mechanism of LID onset. To account for more complex (non-linear) relations of these variables with LID risk, we built a survival random forest (SRF) algorithm including all the covariates mentioned above. Results: Among tested variables (N = 460 case-complete, 211 LID events; total follow-up 31,361 person-months, median 61 months), disease duration showed significant association (p < 0.005), with 6 (3–8)% decrease of LID risk per additional YOD. Other nominally significant associations were observed for gender—with women showing a 39 (5–82)% higher risk of LID—and AAO, with 2 (0.3–3)% decrease of risk for each year increase of PD onset. The SRF algorithm confirmed YOD as the most prominent feature influencing LID risk, with a variable importance of about 8% in the model. In genetic models, no statistically significant effects on incident LID risk was observed. Conclusions: This evidence supports a protective effect of late PD onset and gender (men) against LID risk and suggests a new independent protective factor, YOD. Moreover, it underlines the importance of personalized therapeutic protocols for PD patients in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Teresa Esposito
- IRCCS Neuromed, Pozzilli, Italy.,Institute of Genetics and Biophysics, CNR, Naples, Italy
| |
Collapse
|
44
|
Nakajima T, Suzuki Y, Miyaue N. Successful Management of Parkinson's Disease Dyskinesia During Local Anesthesia With Dexmedetomidine. Cureus 2021; 13:e13739. [PMID: 33842117 PMCID: PMC8020600 DOI: 10.7759/cureus.13739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Dyskinesia is among the most problematic issues for local anaesthesia management in Parkinson's disease (PD) patients. We present a case of a patient with PD who underwent spinal anaesthesia while utilizing dexmedetomidine (DEX) to reduce dyskinesia during urological surgery. DEX stimulates α₂-adrenergic receptors, which works to reduce the dyskinesia in PD patients.
Collapse
Affiliation(s)
- Takamichi Nakajima
- Center for Medical Education and Training, Saiseikai Matsuyama Hospital, Matsuyama, JPN
| | - Yasuyuki Suzuki
- Anesthesiology, Saiseikai Matsuyama Hospital, Matsuyama, JPN
| | | |
Collapse
|
45
|
Lee JW, Song YS, Kim H, Ku BD, Lee WW. Patients with scans without evidence of dopaminergic deficit (SWEDD) do not have early Parkinson's disease: Analysis of the PPMI data. PLoS One 2021; 16:e0246881. [PMID: 33566871 PMCID: PMC7875405 DOI: 10.1371/journal.pone.0246881] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/27/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND To evaluate whether patients with scans without evidence of dopaminergic deficit (SWEDD) have early Parkinson's disease (PD). METHODS The clinical characteristics, striatal specific binding ratios (SBRs), and the indices of I-123 FP-CIT SPECT images of 50 SWEDD patients, 304 PD patients, and 141 healthy controls were acquired from the Parkinson's Progression Markers Initiative (PPMI) data and evaluated during a 2-year clinical follow-up period. RESULTS Of the 50 subjects with SWEDD, PD was confirmed in 13 subjects (the PD-SWEDD group), while the remaining 37 subjects had other diseases (the Other-SWEDD group). Striatal SBR values and striatal asymmetry indices of the PD group were significantly different with those of the PD-SWEDD and Other-SWEDD groups at both baseline and after 2 years (p < 0.001). Putaminal SBR values of the PD-SWEDD group were significantly decreased after 2 years (p < 0.05). There was no difference of the SBR values between baseline and after 2 years in the Other-SWEDD group. A baseline MDS-UPDRS III score matched comparison of the PD and PD-SWEDD group was done due to the large difference of the subject numbers. Striatal SBR values and striatal asymmetry indices were significantly different (p < 0.001) between the two groups at both baseline and after 2 years, but there were no significant difference with respect to the MDS-UPDRS III scores after 2 years between the two groups. CONCLUSION The different SBR values and asymmetry indices between the PD and PD-SWEDD groups at baseline and after 2 years indicate that SWEDD may not be early PD, but rather a different disease entity.
Collapse
Affiliation(s)
- Jeong Won Lee
- Department of Nuclear Medicine, Catholic Kwandong University College of Medicine, International St. Mary’s Hospital, Incheon, Republic of Korea
| | - Yoo Sung Song
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyeyun Kim
- Department of Neurology, Catholic Kwandong University College of Medicine, International St. Mary’s Hospital, Incheon, Republic of Korea
| | - Bon D. Ku
- Department of Neurology, Catholic Kwandong University College of Medicine, International St. Mary’s Hospital, Incheon, Republic of Korea
| | - Won Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
46
|
Julien C, Hache G, Dulac M, Dubrou C, Castelnovo G, Giordana C, Azulay JP, Fluchère F. The clinical meaning of levodopa equivalent daily dose in Parkinson's disease. Fundam Clin Pharmacol 2021; 35:620-630. [PMID: 33458868 DOI: 10.1111/fcp.12646] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/14/2020] [Accepted: 01/12/2021] [Indexed: 12/30/2022]
Abstract
Levodopa (L-dopa) remains the basis of pharmacological treatment of Parkinson's disease (PD). However, L-dopa therapy is associated with the development of complications and presents major challenges in the long-term treatment. Thus, other medications may be suggested to delay and/or reduce the doses of L-dopa in order to prevent complications. The interpretation of treatment evolution reported in clinical trials on PD may be tricky, especially due to some variability in medications and dose regimens. Some authors have suggested a conversion factor to generate a total L-dopa equivalent daily dose (LEDD), calculated as a sum of each parkinsonian medication. Therefore, LEDD provides an artificial summary of the total daily medication a patient is receiving, and to date, there is no report focusing on the clinical interpretation of this parameter. Thus, based on a 3-year, multi-center retrospective study assessing the impact of second-line therapy initiation on LEDD in PD patients, the aim of our article was to discuss LEDD as a quantitative outcome to estimate the impact of second-line therapies on medication regimens; and in the second part of the discussion, to provide a narrative review of the clinical outcomes associated with LEDD in the literature.
Collapse
Affiliation(s)
- Charlotte Julien
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Guillaume Hache
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Morgane Dulac
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Cléa Dubrou
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Giovanni Castelnovo
- Centre Hospitalier Universitaire de Nimes, Service de Neurologie, Nîmes, France
| | - Caroline Giordana
- Centre Hospitalier Universitaire de Nice, Hôpital Pasteur 2, Service de Neurologie, Nice, France
| | - Jean-Philippe Azulay
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Neurologie pathologies du mouvement, Centre expert et de coordination interrégional pour la maladie de Parkinson, Marseille, France
| | - Frédérique Fluchère
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Neurologie pathologies du mouvement, Centre expert et de coordination interrégional pour la maladie de Parkinson, Marseille, France
| |
Collapse
|
47
|
Luca A, Monastero R, Baschi R, Cicero CE, Mostile G, Davì M, Restivo V, Zappia M, Nicoletti A. Cognitive impairment and levodopa induced dyskinesia in Parkinson's disease: a longitudinal study from the PACOS cohort. Sci Rep 2021; 11:867. [PMID: 33441571 PMCID: PMC7806828 DOI: 10.1038/s41598-020-79110-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/02/2020] [Indexed: 11/09/2022] Open
Abstract
Aim of the study was to evaluate possible associations between cognitive dysfunctions and development of Levodopa Induced Dyskinesia (LID). PD patients from the Parkinson’s disease Cognitive impairment Study cohort who underwent a baseline and follow-up neuropsychological evaluations were enrolled. Mild Cognitive Impairment (PD-MCI) was diagnosed according to MDS level II criteria. The following cognitive domains were evaluated: episodic memory, attention, executive function, visuo-spatial function and language. A domain was considered as impaired when the subject scored 2 standard deviation below normality cut-off values in at least one test for each domain. Levodopa equivalent dose, UPDRS-ME and LID were recorded at baseline and follow-up. To identify possible neuropsychological predictors associated with the probability of LID development at follow-up, Cox proportional-hazards regression model was used. Out of 139 PD patients enrolled (87 men, mean age 65.7 ± 9.4), 18 (12.9%) were dyskinetic at baseline. Out of 121 patients non-dyskinetic at baseline, 22 (18.1%) developed LID at follow-up. The impairment of the attention and executive domains strongly predicted the development of LID (HR 4.45;95%CI 1.49–13.23 and HR 3.46; 95%CI 1.26–9.48 respectively). Impairment of the attention and executive domains increased the risk of dyskinesia reflecting the alteration of common cortical network.
Collapse
Affiliation(s)
- Antonina Luca
- Department of Medical, Surgical Sciences and Advanced Technologies, GF Ingrassia, Neurologic Unit, AOU "Policlinico-Vittorio Emanuele", University of Catania, Via Santa Sofia n.78, 95100, Catania, Sicily, Italy
| | - Roberto Monastero
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Via G. La Loggia n.1, Palermo, Sicily, Italy.
| | - Roberta Baschi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Via G. La Loggia n.1, Palermo, Sicily, Italy
| | - Calogero Edoardo Cicero
- Department of Medical, Surgical Sciences and Advanced Technologies, GF Ingrassia, Neurologic Unit, AOU "Policlinico-Vittorio Emanuele", University of Catania, Via Santa Sofia n.78, 95100, Catania, Sicily, Italy
| | - Giovanni Mostile
- Department of Medical, Surgical Sciences and Advanced Technologies, GF Ingrassia, Neurologic Unit, AOU "Policlinico-Vittorio Emanuele", University of Catania, Via Santa Sofia n.78, 95100, Catania, Sicily, Italy
| | - Marco Davì
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Via G. La Loggia n.1, Palermo, Sicily, Italy
| | - Vincenzo Restivo
- Department of Sciences for Health Promotion and Mother-Child Care, University of Palermo, Palermo, Italy
| | - Mario Zappia
- Department of Medical, Surgical Sciences and Advanced Technologies, GF Ingrassia, Neurologic Unit, AOU "Policlinico-Vittorio Emanuele", University of Catania, Via Santa Sofia n.78, 95100, Catania, Sicily, Italy
| | - Alessandra Nicoletti
- Department of Medical, Surgical Sciences and Advanced Technologies, GF Ingrassia, Neurologic Unit, AOU "Policlinico-Vittorio Emanuele", University of Catania, Via Santa Sofia n.78, 95100, Catania, Sicily, Italy.
| |
Collapse
|
48
|
Hinkle JT, Perepezko K, Gonzalez LL, Mills KA, Pontone GM. Apathy and Anxiety in De Novo Parkinson's Disease Predict the Severity of Motor Complications. Mov Disord Clin Pract 2021; 8:76-84. [PMID: 33426161 DOI: 10.1002/mdc3.13117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/09/2020] [Accepted: 10/27/2020] [Indexed: 11/11/2022] Open
Abstract
Background Neuropsychiatric and affective symptoms are prevalent in prodromal and clinical Parkinson's disease (PD). Some evidence suggests that they may also signify risk for motor complications (motor fluctuations and dyskinesias) of dopamine replacement therapy (DRT). Objective To determine whether neuropsychiatric symptoms present in de novo PD (ie, before DRT initiation) predict the severity of eventual motor complications of DRT. Methods We used clinical, demographic, neurobehavioral, and neuroimaging data from the Parkinson's Progression Markers Initiative (PPMI), a multicenter observational PD study. Participants were unmedicated at enrollment and 361 initiated DRT during PPMI follow-up. We used Cox proportional hazard and multivariate ordinal mixed-effects regression models to evaluate the relationship between baseline neuropsychiatric symptoms and motor complications as measured by the Movement Disorders Society-revised Unified Parkinson's Disease Rating Scale (MDS-UPDRS). Results The cumulative incidences of dyskinesias and motor fluctuations during follow-up (6.0 ± 1.5 years) were 34.3% and 59.9%, respectively. Both apathy and high trait-anxiety (top quartile) conveyed over two-fold increases in hazard for dyskinesia onset and for adverse impact on activities of daily living caused by both dyskinesias and motor fluctuations. The longitudinal severity of motor fluctuations and dyskinesias was significantly predicted by baseline trait-anxiety and apathy, but not depression. Models were adjusted for dimensionally related symptoms (eg autonomic dysfunction) and potential confounding variables (eg DRT dose). Conclusions Apathy and anxiety levels in de novo PD may be neuropsychiatric biomarkers of vulnerability to earlier and more disabling motor complications of DRT.
Collapse
Affiliation(s)
- Jared T Hinkle
- Medical Scientist Training Program Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Kate Perepezko
- Department of Psychiatry Johns Hopkins University School of Medicine Baltimore Maryland USA.,Department of Mental Health Johns Hopkins Bloomberg School of Public Health Baltimore Maryland USA
| | - Lorenzo L Gonzalez
- Department of Psychiatry Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Kelly A Mills
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Gregory M Pontone
- Department of Psychiatry Johns Hopkins University School of Medicine Baltimore Maryland USA.,Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA
| |
Collapse
|
49
|
Patricio F, Morales-Andrade AA, Patricio-Martínez A, Limón ID. Cannabidiol as a Therapeutic Target: Evidence of its Neuroprotective and Neuromodulatory Function in Parkinson's Disease. Front Pharmacol 2020; 11:595635. [PMID: 33384602 PMCID: PMC7770114 DOI: 10.3389/fphar.2020.595635] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
The phytocannabinoids of Cannabis sativa L. have, since ancient times, been proposed as a pharmacological alternative for treating various central nervous system (CNS) disorders. Interestingly, cannabinoid receptors (CBRs) are highly expressed in the basal ganglia (BG) circuit of both animals and humans. The BG are subcortical structures that regulate the initiation, execution, and orientation of movement. CBRs regulate dopaminergic transmission in the nigro-striatal pathway and, thus, the BG circuit also. The functioning of the BG is affected in pathologies related to movement disorders, especially those occurring in Parkinson’s disease (PD), which produces motor and non-motor symptoms that involving GABAergic, glutamatergic, and dopaminergic neural networks. To date, the most effective medication for PD is levodopa (l-DOPA); however, long-term levodopa treatment causes a type of long-term dyskinesias, l-DOPA-induced dyskinesias (LIDs). With neuromodulation offering a novel treatment strategy for PD patients, research has focused on the endocannabinoid system (ECS), as it participates in the physiological neuromodulation of the BG in order to control movement. CBRs have been shown to inhibit neurotransmitter release, while endocannabinoids (eCBs) play a key role in the synaptic regulation of the BG. In the past decade, cannabidiol (CBD), a non-psychotropic phytocannabinoid, has been shown to have compensatory effects both on the ECS and as a neuromodulator and neuroprotector in models such as 6-hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and reserpine, as well as other PD models. Although the CBD-induced neuroprotection observed in animal models of PD has been attributed to the activation of the CB1 receptor, recent research conducted at a molecular level has proposed that CBD is capable of activating other receptors, such as CB2 and the TRPV-1 receptor, both of which are expressed in the dopaminergic neurons of the nigro-striatal pathway. These findings open new lines of scientific inquiry into the effects of CBD at the level of neural communication. Cannabidiol activates the PPARγ, GPR55, GPR3, GPR6, GPR12, and GPR18 receptors, causing a variety of biochemical, molecular, and behavioral effects due to the broad range of receptors it activates in the CNS. Given the low number of pharmacological treatment alternatives for PD currently available, the search for molecules with the therapeutic potential to improve neuronal communication is crucial. Therefore, the investigation of CBD and the mechanisms involved in its function is required in order to ascertain whether receptor activation could be a treatment alternative for both PD and LID.
Collapse
Affiliation(s)
- Felipe Patricio
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alan Axel Morales-Andrade
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.,Facultad De Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio De Neurofarmacología, Facultad De Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
50
|
Dos Santos EUD, da Silva IIFG, Asano AGC, Asano NMJ, De Mascena Diniz Maia M, de Souza PRE. Pharmacogenetic profile and the development of the dyskinesia induced by levodopa-therapy in Parkinson's disease patients: a population-based cohort study. Mol Biol Rep 2020; 47:8997-9004. [PMID: 33151475 DOI: 10.1007/s11033-020-05956-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/28/2020] [Indexed: 12/21/2022]
Abstract
Levodopa-induced dyskinesia (LID) is an adverse effect that negatively impacts the quality of life of patients with Parkinson's disease (PD). Studies report that genetic variations in the genes of the pharmacogenetic pathway of the levodopa (L-DOPA) might be associated with LID development. The goal of the present study was to investigate a possible influence of functional genetic variants in the DRD1 (rs4532), DRD2 (rs1800497), DAT1 (rs28363170), and COMT (rs4680) genes with LID development. A total of 220 patients with idiopathic PD were enrolled. The genotyping for DRD1 (rs4532), DRD2 (rs1800497), DAT1 (rs28363170), and COMT (rs4680) polymorphisms were performed using Restriction Fragment Length Polymorphism (PCR-RFLP). Univariate and multivariate analyses were performed to assess the association of these polymorphisms and risk factors with LID development. Multivariate Cox regression analysis showed increased risk to LID development for both Levodopa Dose Equivalency (LED) (Hazard ratios (HR) = 1.001; 95% CI 1.00-1.01; p = 0.009) and individuals carrying the COMT L/L genotype (HR = 2.974; 95% CI 1.12-7.83; p = 0.010). Furthermore, when performed a Cox regression analysis adjusted for a total LED, we observed that the genotype COMT L/L had a 3.84-fold increased risk for LID development (HR = 3.841; 95% CI 1.29-11.37; p = 0.012). Our results suggest that before treating LID in PD patients, it is important to take into consideration genetic variant in the COMT gene, since COMT LL genotype may increase the risk for LID development.
Collapse
Affiliation(s)
- Erinaldo Ubirajara Damasceno Dos Santos
- Graduate Program in Applied Cellular and Molecular Biology, University of Pernambuco (UPE), Rua Dom Manuel de Medeiros, S/N -Dois Irmãos, CEP:52171-900, Recife, PE, Brazil
| | | | - Amdore Guescel C Asano
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Pernambuco (UFPE), Recife, PE, Brazil.,Pro-Parkinson Program of the Clinical Hospital of the Federal University of Pernambuco Recife (HC/UFPE), Recife, PE, Brazil
| | - Nadja Maria Jorge Asano
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Pernambuco (UFPE), Recife, PE, Brazil.,Pro-Parkinson Program of the Clinical Hospital of the Federal University of Pernambuco Recife (HC/UFPE), Recife, PE, Brazil
| | | | - Paulo Roberto Eleutério de Souza
- Graduate Program in Applied Cellular and Molecular Biology, University of Pernambuco (UPE), Rua Dom Manuel de Medeiros, S/N -Dois Irmãos, CEP:52171-900, Recife, PE, Brazil. .,Graduate Program in Genetics, Federal University of Pernambuco (UFPE), Recife, PE, Brazil. .,Department of Biology, Federal Rural University of Pernambuco (UFRPE), Recife, PE, Brazil.
| |
Collapse
|