1
|
Guidetti M, Bocci T, De Pedro Del Álamo M, Deuschl G, Fasano A, Martinez-Fernandez R, Gasca-Salas C, Hamani C, Krauss JK, Kühn AA, Limousin P, Little S, Lozano AM, Maiorana NV, Marceglia S, Okun MS, Oliveri S, Ostrem JL, Scelzo E, Schnitzler A, Starr PA, Temel Y, Timmermann L, Tinkhauser G, Visser-Vandewalle V, Volkmann J, Priori A. Will adaptive deep brain stimulation for Parkinson's disease become a real option soon? A Delphi consensus study. NPJ Parkinsons Dis 2025; 11:110. [PMID: 40325017 PMCID: PMC12052990 DOI: 10.1038/s41531-025-00974-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/19/2025] [Indexed: 05/07/2025] Open
Abstract
While conventional deep brain stimulation (cDBS) treatment delivers continuous electrical stimuli, new adaptive DBS (aDBS) technology provides dynamic symptom-related stimulation. Research data are promising, and devices are already available, but are we ready for it? We asked leading DBS experts worldwide (n = 21) to discuss a research agenda for aDBS research in the near future to allow full adoption. A 5-point Likert scale questionnaire, along with a Delphi method, was employed. In the next 10 years, aDBS will be clinical routine, but research is needed to define which patients would benefit more from the treatment; second, implantation and programming procedures should be simplified to allow actual generalized adoption; third, new adaptive algorithms, and the integration of aDBS paradigm with new technologies, will improve control of more complex symptoms. Since the next years will be crucial for aDBS implementation, the research should focus on improving precision and making programming procedures more accessible.
Collapse
Affiliation(s)
- Matteo Guidetti
- Department of Health Sciences, "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, Milan, Italy
| | - Tommaso Bocci
- Department of Health Sciences, "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, Milan, Italy
- Clinical Neurology Unit, Department of Health Sciences, "Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo", University of Milan, Milan, Italy
| | | | - Guenther Deuschl
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Christian Albrechts-University of Kiel Kiel Germany, Kiel, Germany
| | - Alfonso Fasano
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- CRANIA Center for Advancing Neurotechnological Innovation to Application, University of Toronto, Toronto, ON, Canada
- KITE, University Health Network, Toronto, ON, Canada
- Edmond J. Safra Program in Parkinson's Disease Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - Raul Martinez-Fernandez
- HM CINAC, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto Carlos III, CIBERNED, Madrid, Spain
| | - Carmen Gasca-Salas
- HM CINAC, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto Carlos III, CIBERNED, Madrid, Spain
| | - Clement Hamani
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Harquail Centre for Neuromodulation, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Andrea A Kühn
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität, Berlin, Germany
- NeuroCure, Exzellenzcluster, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZNE, German Center for Neurodegenerative Diseases, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Patricia Limousin
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, UK
| | - Simon Little
- Movement Disorders and Neuromodulation Centre, University of California San Francisco, San Francisco, CA, USA
| | - Andres M Lozano
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- CRANIA Center for Advancing Neurotechnological Innovation to Application, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Natale V Maiorana
- Department of Health Sciences, "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, Milan, Italy
| | - Sara Marceglia
- Department of Health Sciences, "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, Milan, Italy.
| | - Michael S Okun
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Serena Oliveri
- Department of Health Sciences, "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, Milan, Italy
- Clinical Neurology Unit, Department of Health Sciences, "Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo", University of Milan, Milan, Italy
| | - Jill L Ostrem
- Movement Disorders and Neuromodulation Centre, University of California San Francisco, San Francisco, CA, USA
| | - Emma Scelzo
- Clinical Neurology Unit, Department of Health Sciences, "Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo", University of Milan, Milan, Italy
| | - Alfons Schnitzler
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Department of Neurology, Center for Movement Disorders and Neuromodulation, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Philip A Starr
- UCSF Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- UCSF Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lars Timmermann
- Department of Neurology, University Hospital of Marburg, Marburg, Germany
| | - Gerd Tinkhauser
- Department of Neurology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Veerle Visser-Vandewalle
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Alberto Priori
- Department of Health Sciences, "Aldo Ravelli" Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, Milan, Italy
- Clinical Neurology Unit, Department of Health Sciences, "Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo", University of Milan, Milan, Italy
| |
Collapse
|
2
|
Du D, Fu W, Su S, Mao X, Yang L, Xu M, Yuan Y, Gao Y, Geng Z, Chen Y, Zhao M, Fu Y, Yin F, Han H. Remote Regulation of Molecular Diffusion in Extracellular Space of Parkinson's Disease Rat Model by Subthalamic Nucleus Deep Brain Stimulation. CYBORG AND BIONIC SYSTEMS 2025; 6:0218. [PMID: 40190716 PMCID: PMC11969791 DOI: 10.34133/cbsystems.0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/30/2024] [Accepted: 12/29/2024] [Indexed: 04/09/2025] Open
Abstract
Subthalamic nucleus deep brain stimulation (STN-DBS) is an effective therapy for Parkinson's disease (PD). However, the therapeutic mechanisms remain incompletely understood, particularly regarding the extracellular space (ECS), a critical microenvironment where molecular diffusion and interstitial fluid (ISF) dynamics are essential for neural function. This study aims to explore the regulatory mechanisms of the ECS in the substantia nigra (SN) of PD rats following STN-DBS. To evaluate whether STN-DBS can modulate ECS diffusion and drainage, we conducted quantitative measurements using a tracer-based magnetic resonance imaging. Our findings indicated that, compared to the PD group, STN-DBS treatment resulted in a decreased diffusion coefficient (D*), shorted half-life (T 1/2), and increased clearance coefficient (k') in the SN. To investigate the mechanisms underlying these changes in molecular diffusion, we employed enzyme-linked immunosorbent assay (ELISA), Western blotting (WB), and microdialysis techniques. The results revealed that STN-DBS led to an increase in hyaluronic acid content, elevated expression of excitatory amino acid transporter 2 (EAAT2), and a reduction in extracellular glutamate concentration. Additionally, to further elucidate the mechanisms influencing ISF drainage, we employed immunofluorescence and immunohistochemical techniques for staining aquaporin-4 (AQP-4) and α-synuclein. The results demonstrated that STN-DBS restored the expression of AQP-4 while decreasing the expression of α-synuclein. In conclusion, our findings suggest that STN-DBS improves PD symptoms by modifying the ECS and enhancing ISF drainage in the SN regions. These results offer new insights into the mechanisms and long-term outcomes of DBS in ECS, paving the way for precision therapies.
Collapse
Affiliation(s)
- Dan Du
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao 066000, China
| | - Wanyi Fu
- Department of Electronic Engineering,
Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology,
Peking University Third Hospital, Beijing 100191, China
| | - Shaoyi Su
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Xin Mao
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology,
Peking University Third Hospital, Beijing 100191, China
| | - Liu Yang
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
| | - Meng Xu
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
| | - Yi Yuan
- School of Electrical Engineering,
Yanshan University, Qinhuangdao 066004, China
| | - Yajuan Gao
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology,
Peking University Third Hospital, Beijing 100191, China
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
- National Medical Products Administration Key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing 100191, China
| | - Ziyao Geng
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology,
Peking University Third Hospital, Beijing 100191, China
| | - Yanjing Chen
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
| | - Mingming Zhao
- Department of Neurosurgery, Aerospace Center Hospital, Beijing 100049, China
| | - Yu Fu
- Department of Neurology,
Peking University Third Hospital, Beijing 100191, China
| | - Feng Yin
- Department of Neurosurgery, Aerospace Center Hospital, Beijing 100049, China
| | - Hongbin Han
- Department of Radiology,
Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology,
Peking University Third Hospital, Beijing 100191, China
- Institute of Medical Technology,
Peking University Health Science Center, Beijing 100191, China
- National Medical Products Administration Key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing 100191, China
| |
Collapse
|
3
|
Li Y, Nie Y, Li X, Cheng X, Zhu G, Zhang J, Quan Z, Wang S. Closed-Loop Deep Brain Stimulation Platform for Translational Research. Neuromodulation 2025; 28:464-471. [PMID: 39674932 DOI: 10.1016/j.neurom.2024.10.012] [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: 07/16/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE This study aims to facilitate the translation of innovative closed-loop deep brain stimulation (DBS) strategies from theory to practice by establishing a research platform. The platform addresses the challenges of real-time stimulation artifact removal, low-latency feedback stimulation, and rapid translation from animal to clinical experiments. MATERIALS AND METHODS The platform comprises hardware for neural sensing and stimulation, a closed-loop software framework for real-time data streaming and computation, and an algorithm library for implementing closed-loop DBS strategies. The platform integrates hardware for both animal and clinical research. The closed-loop software framework handles the entire closed-loop stimulation, including data streaming, stimulation artifact removal, preprocessing, a closed-loop stimulation strategy, and stimulation control. It provides a unified programming interface for both C/C++ and Python, enabling secondary development to integrate new closed-loop stimulation strategies. Additionally, the platform includes an algorithm library with signal processing and machine learning methods to facilitate the development of new closed-loop DBS strategies. RESULTS The platform can achieve low-latency feedback stimulation control with response times of 6.23 ± 0.85 ms and 6.95 ± 1.11 ms for animal and clinical experiments, respectively. It effectively removed stimulation artifacts and demonstrated flexibility in implementing new closed-loop DBS algorithms. The platform has integrated several typical closed-loop protocols, including threshold-adaptive DBS, amplitude-modulation DBS, dual-threshold DBS and neural state-dependent DBS. CONCLUSIONS This work provides a research tool for rapidly deploying innovative closed-loop strategies for translational research in both animal and clinical studies. The platform's capabilities in real-time data processing and low-latency control represent a significant advancement in translational DBS research, with potential implications for the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Yan Li
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yingnan Nie
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xiao Li
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xi Cheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Guanyu Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhaoyu Quan
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Shouyan Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China; Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Academy for Engineering and Technology, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Mirpour K, Pouratian N. Interaction of motor behaviour, cortical oscillations and deep brain stimulation in Parkinson's disease. Brain 2025; 148:886-895. [PMID: 39300838 PMCID: PMC11884658 DOI: 10.1093/brain/awae300] [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: 02/12/2024] [Revised: 08/04/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
Recent progress in the study of Parkinson's disease has highlighted the pivotal role of beta oscillations within the basal ganglia-thalamo-cortical network in modulating motor symptoms. Predominantly manifesting as transient bursts, these beta oscillations are central to the pathophysiology of Parkinson's disease motor symptoms, especially bradykinesia. Our central hypothesis is that increased bursting duration in cortex, coupled with kinematics of movement, disrupts the typical flow of neural information, leading to observable changes in motor behaviour in Parkinson's disease. To explore this hypothesis, we employed an integrative approach, analysing the interplay between moment-to-moment brain dynamics and movement kinematics and the modulation of these relationships by therapeutic deep brain stimulation (DBS). Local field potentials were recorded from the hand motor (M1) and premotor cortical (PM) areas and internal globus pallidus (GPi) in 26 patients with Parkinson's disease undergoing DBS implantation surgery. Participants executed rapid alternating hand movements in 30-s blocks, both with and without therapeutic pallidal stimulation. Behaviourally, the analysis revealed bradykinesia, with hand movement cycle width increasing linearly over time during DBS-OFF blocks. Crucially, there was a moment-to-moment correlation between M1 low beta burst duration and movement cycle width, a relationship that dissipated with therapeutic DBS. Further analyses suggested that high gamma activity correlates with enhanced motor performance with DBS-ON. Regardless of the nature of coupling, DBS's modulation of cortical bursting activity appeared to amplify the brain signals' informational content regarding instantaneous movement changes. Our findings underscore that DBS significantly reshapes the interaction between motor behaviour and neural signals in Parkinson's disease, not only modulating specific bands but also expanding the system's capability to process and relay information for motor control. These insights shed light on the possible network mechanisms underlying DBS therapeutic effects, suggesting a profound impact on both neural and motor domains.
Collapse
Affiliation(s)
- Koorosh Mirpour
- Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nader Pouratian
- Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
5
|
Gobeil M, Guillemette A, Silhadi M, Charbonneau L, Bergeron D, Dominguez‐Vargas A, Dancause N, Jodoin N, Assi E, Amzica F, Obaid S, Fournier‐Gosselin M. Local Field Potential Biomarkers of Non-Motor Symptoms in Parkinson's Disease: Insights From the Subthalamic Nucleus in Deep Brain Stimulation. Eur J Neurosci 2025; 61:e70046. [PMID: 40040306 PMCID: PMC11880747 DOI: 10.1111/ejn.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
Non-motor symptoms can severely affect the quality of life of Parkinson's disease-afflicted patients, with the most common ones being pain, sleep impairments, and neuropsychiatric manifestations. In advanced cases, complex fluctuations of motor and non-motor symptoms can occur despite optimal medication. Research on deep brain stimulation of the subthalamic nucleus suggests that it may provide benefits for treating non-motor symptoms in addition to improving motor symptoms. With recent advancements in deep brain stimulation technology, simultaneous recording of local field potentials and delivery of therapeutic stimulation is possible. This opens new possibilities for better understanding the pathophysiology of non-motor symptoms in Parkinson's disease and for identifying potential electrophysiological biomarkers that accurately represent these symptoms. Specifically, this review aims to highlight potential local field potential biomarkers of non-motor symptoms in the subthalamic nucleus. The main findings indicate that activities in the beta frequency band are associated with nociception and sleep impairments such as insomnia and rapid eye movement sleep behavior disorders. Additionally, activities in the theta and alpha frequency bands seem to reflect neurocognitive manifestations, including depression and impulse control disorders. A better understanding of these biomarkers could improve the clinical management of non-motor symptoms in Parkinson's disease. They hold promise for adjusting deep brain stimulation parameters in open-loop settings and might eventually be applied in closed-loop deep brain stimulation systems, though their true impact remains uncertain.
Collapse
Affiliation(s)
- Marc‐Antoine Gobeil
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
| | - Albert Guillemette
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
| | - Meziane Silhadi
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
| | - Laurence Charbonneau
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
- Department of SurgeryUniversity of MontrealMontrealQuebecCanada
| | - David Bergeron
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
- Department of SurgeryUniversity of MontrealMontrealQuebecCanada
- Department of NeurosciencesUniversity of MontrealMontrealQuebecCanada
| | - Adan‐Ulises Dominguez‐Vargas
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Department of NeurosciencesUniversity of MontrealMontrealQuebecCanada
| | - Numa Dancause
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Department of NeurosciencesUniversity of MontrealMontrealQuebecCanada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l'Apprentissage (CIRCA)MontrealQuebecCanada
| | - Nicolas Jodoin
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
- Department of NeurosciencesUniversity of MontrealMontrealQuebecCanada
- Division of NeurologyCentre Hospitalier de l'Université de Montréal (CHUM)MontrealQuebecCanada
| | - Elie Bou Assi
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
- Department of NeurosciencesUniversity of MontrealMontrealQuebecCanada
| | - Florin Amzica
- Department of NeurosciencesUniversity of MontrealMontrealQuebecCanada
- Department of StomatologyUniversity of MontrealMontrealQuebecCanada
| | - Sami Obaid
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
- Department of SurgeryUniversity of MontrealMontrealQuebecCanada
- Service of Neurosurgery, Centre Hospitalier de l'Université de Montréal (CHUM)MontrealQuebecCanada
| | - Marie‐Pierre Fournier‐Gosselin
- Department of MedicineUniversity of MontrealMontrealQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada
- Department of SurgeryUniversity of MontrealMontrealQuebecCanada
- Service of Neurosurgery, Centre Hospitalier de l'Université de Montréal (CHUM)MontrealQuebecCanada
| |
Collapse
|
6
|
Asadi A, Wiesman AI, Wiest C, Baillet S, Tan H, Muthuraman M. Electrophysiological approaches to informing therapeutic interventions with deep brain stimulation. NPJ Parkinsons Dis 2025; 11:20. [PMID: 39833210 PMCID: PMC11747345 DOI: 10.1038/s41531-024-00847-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Neuromodulation therapy comprises a range of non-destructive and adjustable methods for modulating neural activity using electrical stimulations, chemical agents, or mechanical interventions. Here, we discuss how electrophysiological brain recording and imaging at multiple scales, from cells to large-scale brain networks, contribute to defining the target location and stimulation parameters of neuromodulation, with an emphasis on deep brain stimulation (DBS).
Collapse
Affiliation(s)
- Atefeh Asadi
- Neural Engineering with Signal Analytics and Artificial Intelligence, Department of Neurology, University Clinic Würzburg, Würzburg, Germany.
| | - Alex I Wiesman
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Christoph Wiest
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sylvain Baillet
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Huiling Tan
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Muthuraman Muthuraman
- Neural Engineering with Signal Analytics and Artificial Intelligence, Department of Neurology, University Clinic Würzburg, Würzburg, Germany
- Informatics for Medical Technology, Institute of Computer Science, University Augsburg, Augsburg, Germany
| |
Collapse
|
7
|
Kumar R, Waisberg E, Ong J, Paladugu P, Amiri D, Saintyl J, Yelamanchi J, Nahouraii R, Jagadeesan R, Tavakkoli A. Artificial Intelligence-Based Methodologies for Early Diagnostic Precision and Personalized Therapeutic Strategies in Neuro-Ophthalmic and Neurodegenerative Pathologies. Brain Sci 2024; 14:1266. [PMID: 39766465 PMCID: PMC11674895 DOI: 10.3390/brainsci14121266] [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: 11/20/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Advancements in neuroimaging, particularly diffusion magnetic resonance imaging (MRI) techniques and molecular imaging with positron emission tomography (PET), have significantly enhanced the early detection of biomarkers in neurodegenerative and neuro-ophthalmic disorders. These include Alzheimer's disease, Parkinson's disease, multiple sclerosis, neuromyelitis optica, and myelin oligodendrocyte glycoprotein antibody disease. This review highlights the transformative role of advanced diffusion MRI techniques-Neurite Orientation Dispersion and Density Imaging and Diffusion Kurtosis Imaging-in identifying subtle microstructural changes in the brain and visual pathways that precede clinical symptoms. When integrated with artificial intelligence (AI) algorithms, these techniques achieve unprecedented diagnostic precision, facilitating early detection of neurodegeneration and inflammation. Additionally, next-generation PET tracers targeting misfolded proteins, such as tau and alpha-synuclein, along with inflammatory markers, enhance the visualization and quantification of pathological processes in vivo. Deep learning models, including convolutional neural networks and multimodal transformers, further improve diagnostic accuracy by integrating multimodal imaging data and predicting disease progression. Despite challenges such as technical variability, data privacy concerns, and regulatory barriers, the potential of AI-enhanced neuroimaging to revolutionize early diagnosis and personalized treatment in neurodegenerative and neuro-ophthalmic disorders is immense. This review underscores the importance of ongoing efforts to validate, standardize, and implement these technologies to maximize their clinical impact.
Collapse
Affiliation(s)
- Rahul Kumar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA; (R.K.); (J.S.)
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK;
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St, Ann Arbor, MI 48105, USA
| | - Phani Paladugu
- Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St, Philadelphia, PA 19107, USA;
- Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Dylan Amiri
- Department of Biology, University of Miami, 1301 Memorial Dr, Coral Gables, FL 33146, USA;
- Mecklenburg Neurology Group, 3541 Randolph Rd #301, Charlotte, NC 28211, USA;
| | - Jeremy Saintyl
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1600 NW 10th Ave, Miami, FL 33136, USA; (R.K.); (J.S.)
| | - Jahnavi Yelamanchi
- Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY 11201, USA;
| | - Robert Nahouraii
- Mecklenburg Neurology Group, 3541 Randolph Rd #301, Charlotte, NC 28211, USA;
| | - Ram Jagadeesan
- Whiting School of Engineering, Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA;
| | - Alireza Tavakkoli
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, 1664 N Virginia St, Reno, NV 89557, USA;
| |
Collapse
|
8
|
Cho CH, Huang PJ, Chen MC, Lin CW. Closed-Loop Deep Brain Stimulation With Reinforcement Learning and Neural Simulation. IEEE Trans Neural Syst Rehabil Eng 2024; 32:3615-3624. [PMID: 39302783 DOI: 10.1109/tnsre.2024.3465243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Deep Brain Stimulation (DBS) is effective for movement disorders, particularly Parkinson's disease (PD). However, a closed-loop DBS system using reinforcement learning (RL) for automatic parameter tuning, offering enhanced energy efficiency and the effect of thalamus restoration, is yet to be developed for clinical and commercial applications. In this research, we instantiate a basal ganglia-thalamic (BGT) model and design it as an interactive environment suitable for RL models. Four finely tuned RL agents based on different frameworks, namely Soft Actor-Critic (SAC), Twin Delayed Deep Deterministic Policy Gradient (TD3), Proximal Policy Optimization (PPO), and Advantage Actor-Critic (A2C), are established for further comparison. Within the implemented RL architectures, the optimized TD3 demonstrates a significant 67% reduction in average power dissipation when compared to the open-loop system while preserving the normal response of the simulated BGT circuitry. As a result, our method mitigates thalamic error responses under pathological conditions and prevents overstimulation. In summary, this study introduces a novel approach to implementing an adaptive parameter-tuning closed-loop DBS system. Leveraging the advantages of TD3, our proposed approach holds significant promise for advancing the integration of RL applications into DBS systems, ultimately optimizing therapeutic effects in future clinical trials.
Collapse
|
9
|
Stanslaski S, Summers RLS, Tonder L, Tan Y, Case M, Raike RS, Morelli N, Herrington TM, Beudel M, Ostrem JL, Little S, Almeida L, Ramirez-Zamora A, Fasano A, Hassell T, Mitchell KT, Moro E, Gostkowski M, Sarangmat N, Bronte-Stewart H. Sensing data and methodology from the Adaptive DBS Algorithm for Personalized Therapy in Parkinson's Disease (ADAPT-PD) clinical trial. NPJ Parkinsons Dis 2024; 10:174. [PMID: 39289373 PMCID: PMC11408616 DOI: 10.1038/s41531-024-00772-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
Adaptive deep brain stimulation (aDBS) is an emerging advancement in DBS technology; however, local field potential (LFP) signal rate detection sufficient for aDBS algorithms and the methods to set-up aDBS have yet to be defined. Here we summarize sensing data and aDBS programming steps associated with the ongoing Adaptive DBS Algorithm for Personalized Therapy in Parkinson's Disease (ADAPT-PD) pivotal trial (NCT04547712). Sixty-eight patients were enrolled with either subthalamic nucleus or globus pallidus internus DBS leads connected to a Medtronic PerceptTM PC neurostimulator. During the enrollment and screening procedures, a LFP (8-30 Hz, ≥1.2 µVp) control signal was identified by clinicians in 84.8% of patients on medication (65% bilateral signal), and in 92% of patients off medication (78% bilateral signal). The ADAPT-PD trial sensing data indicate a high LFP signal presence in both on and off medication states of these patients, with bilateral signal in the majority, regardless of PD phenotype.
Collapse
Affiliation(s)
- Scott Stanslaski
- Medtronic Neuromodulation, Medtronic, Minneapolis, Minnesota, USA.
| | | | - Lisa Tonder
- Medtronic Neuromodulation, Medtronic, Minneapolis, Minnesota, USA
| | - Ye Tan
- Medtronic Neuromodulation, Medtronic, Minneapolis, Minnesota, USA
| | - Michelle Case
- Medtronic Neuromodulation, Medtronic, Minneapolis, Minnesota, USA
| | - Robert S Raike
- Medtronic Neuromodulation, Medtronic, Minneapolis, Minnesota, USA
| | - Nathan Morelli
- Medtronic Neuromodulation, Medtronic, Minneapolis, Minnesota, USA
| | | | - Martijn Beudel
- Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jill L Ostrem
- Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Simon Little
- Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Leonardo Almeida
- Department of Neurology, University of Minnesota, Minneapolis, USA
| | - Adolfo Ramirez-Zamora
- Department of Neurology, Shands at University of Florida, University of Florida, Gainesville, USA
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, University of Toronto, Toronto, ON, Canada
- Krembil Brain Institute, University of Toronto, Toronto, ON, Canada
| | - Travis Hassell
- Department of Neurology, Vanderbilt University Medical Center, Nashville, USA
| | - Kyle T Mitchell
- Duke University Movement Disorders Center, Duke University, Durham, USA
| | - Elena Moro
- Grenoble Alpes University, Division of Neurology, Grenoble Institute of Neuroscience, CHU of Grenoble, Grenoble, France
| | - Michal Gostkowski
- Center for Neurological Restoration, Cleveland Clinic Foundation, Cleveland, USA
| | | | - Helen Bronte-Stewart
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA
| |
Collapse
|
10
|
Fasano A, Mure H, Oyama G, Murase N, Witt T, Higuchi Y, Singer A, Sannelli C, Morelli N. Subthalamic nucleus local field potential stability in patients with Parkinson's disease. Neurobiol Dis 2024; 199:106589. [PMID: 38969232 DOI: 10.1016/j.nbd.2024.106589] [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: 03/05/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Despite the large body of work on local field potentials (LFPs), a measure of oscillatory activity in patients with Parkinson's disease (PD), the longitudinal evolution of LFPs is less explored. OBJECTIVE To determine LFP fluctuations collected in clinical settings in patients with PD and STN deep brain stimulation (DBS). METHODS Twenty-two STN-DBS patients (age: 67.6 ± 8.3 years; 9 females; disease duration: 10.3 ± 4.5 years) completed bilateral LFP recordings over three visits in the OFF-stimulation setting. Peak and band power measures were calculated from each recording. RESULTS After bilateral LFP recordings, at least one peak was detected in 18 (81.8%), 20 (90.9%), and 22 (100%) patients at visit 1, 2, and 3, respectively. No significant differences were seen in primary peak amplitude (F = 2.91, p = 0.060) over time. Amplitude of the second largest peak (F = 5.49, p = 0.006) and low-beta (F = 6.89, p = 0.002), high-beta (F = 13.23, p < 0.001), and gamma (F = 12.71, p < 0.001) band power demonstrated a significant effect of time. Post hoc comparisons determined low-beta power (Visit 1-Visit 2: t = 3.59, p = 0.002; Visit 1-Visit 3: t = 2.61, p = 0.031), high-beta (Visit 1-Visit 2: t = 4.64, p < 0.001; Visit 1-Visit 3: t = 4.23, p < 0.001) and gamma band power (Visit 1-Visit 2: t = 4.65, p < 0.001; Visit 1-Visit 3: t = 4.00, p < 0.001) were significantly increased from visit 1 recordings to both follow-up visits. CONCLUSION Our results provide substantial evidence that LFP can reliably be detected across multiple real-world clinical visits in patients with STN-DBS for PD. Moreover, it provides insights on the evolution of these LFPs.
Collapse
Affiliation(s)
- Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Toronto, Canada; Division of Neurology, University of Toronto, Toronto, Canada; Krembil Brain Institute, University Health Network, Toronto, Canada; Center for Advancing Neurotechnological Innovation to Application, Toronto, Canada.
| | - Hideo Mure
- Center for Neuromodulation, Department of Neurosurgery, Kurashiki Heisei Hospital, Kurashiki, Japan
| | - Genko Oyama
- Department of Neurology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Nagako Murase
- Department of Neurology, National Hospital Organization Nara Medical Center, Nara, Japan
| | - Thomas Witt
- Department of Neurosurgery, Indiana University Medical Center, Indianapolis, IN, USA
| | - Yoshinori Higuchi
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University Hospital, Chiba, Japan
| | - Alexa Singer
- Brain Modulation Business, Neuromodulation Operating Unit, Medtronic PLC, Minneapolis, MN, USA
| | - Claudia Sannelli
- Brain Modulation Business, Neuromodulation Operating Unit, Medtronic PLC, Minneapolis, MN, USA
| | - Nathan Morelli
- Brain Modulation Business, Neuromodulation Operating Unit, Medtronic PLC, Minneapolis, MN, USA
| |
Collapse
|
11
|
Rohr-Fukuma M, Stieglitz LH, Bujan B, Jedrysiak P, Oertel MF, Salzmann L, Baumann CR, Imbach LL, Gassert R, Bichsel O. Neurofeedback-enabled beta power control with a fully implanted DBS system in patients with Parkinson's disease. Clin Neurophysiol 2024; 165:1-15. [PMID: 38941959 DOI: 10.1016/j.clinph.2024.06.001] [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: 09/13/2023] [Revised: 04/18/2024] [Accepted: 06/03/2024] [Indexed: 06/30/2024]
Abstract
OBJECTIVE Parkinsonian motor symptoms are linked to pathologically increased beta oscillations in the basal ganglia. Studies with externalised deep brain stimulation electrodes showed that Parkinson patients were able to rapidly gain control over these pathological basal ganglia signals through neurofeedback. Studies with fully implanted deep brain stimulation systems duplicating these promising results are required to grant transferability to daily application. METHODS In this study, seven patients with idiopathic Parkinson's disease and one with familial Parkinson's disease were included. In a postoperative setting, beta oscillations from the subthalamic nucleus were recorded with a fully implanted deep brain stimulation system and converted to a real-time visual feedback signal. Participants were instructed to perform bidirectional neurofeedback tasks with the aim to modulate these oscillations. RESULTS While receiving regular medication and deep brain stimulation, participants were able to significantly improve their neurofeedback ability and achieved a significant decrease of subthalamic beta power (median reduction of 31% in the final neurofeedback block). CONCLUSION We could demonstrate that a fully implanted deep brain stimulation system can provide visual neurofeedback enabling patients with Parkinson's disease to rapidly control pathological subthalamic beta oscillations. SIGNIFICANCE Fully-implanted DBS electrode-guided neurofeedback is feasible and can now be explored over extended timespans.
Collapse
Affiliation(s)
- Manabu Rohr-Fukuma
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Switzerland; Clinical Neuroscience Centre, University Hospital Zurich, University of Zurich, Switzerland
| | - Lennart H Stieglitz
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Switzerland; Clinical Neuroscience Centre, University Hospital Zurich, University of Zurich, Switzerland
| | | | | | - Markus F Oertel
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Switzerland; Clinical Neuroscience Centre, University Hospital Zurich, University of Zurich, Switzerland
| | - Lena Salzmann
- Rehabilitation Engineering Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Christian R Baumann
- Clinical Neuroscience Centre, University Hospital Zurich, University of Zurich, Switzerland; Department of Neurology, University Hospital Zurich, University of Zurich, Switzerland
| | | | - Roger Gassert
- Rehabilitation Engineering Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Oliver Bichsel
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Switzerland; Clinical Neuroscience Centre, University Hospital Zurich, University of Zurich, Switzerland; Rehabilitation Engineering Laboratory, Department of Health Sciences and Technology, ETH Zurich, Switzerland.
| |
Collapse
|
12
|
Guidetti M, Bocci T, De Pedro Del Álamo M, Deuschl G, Fasano A, Fernandez RM, Gasca-Salas C, Hamani C, Krauss J, Kühn AA, Limousin P, Little S, Lozano A, Maiorana N, Marceglia S, Okun M, Oliveri S, Ostrem JL, Scelzo E, Schnitzler A, Starr P, Temel Y, Timmermann L, Tinkhauser G, Visser-Vandewalle V, Volkmann J, Priori A. Adaptive Deep Brain Stimulation in Parkinson's Disease: A Delphi Consensus Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.26.24312580. [PMID: 39252901 PMCID: PMC11383503 DOI: 10.1101/2024.08.26.24312580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Importance If history teaches, as cardiac pacing moved from fixed-rate to on-demand delivery in in 80s of the last century, there are high probabilities that closed-loop and adaptive approaches will become, in the next decade, the natural evolution of conventional Deep Brain Stimulation (cDBS). However, while devices for aDBS are already available for clinical use, few data on their clinical application and technological limitations are available so far. In such scenario, gathering the opinion and expertise of leading investigators worldwide would boost and guide practice and research, thus grounding the clinical development of aDBS. Observations We identified clinical and academically experienced DBS clinicians (n=21) to discuss the challenges related to aDBS. A 5-point Likert scale questionnaire along with a Delphi method was employed. 42 questions were submitted to the panel, half of them being related to technical aspects while the other half to clinical aspects of aDBS. Experts agreed that aDBS will become clinical practice in 10 years. In the present scenario, although the panel agreed that aDBS applications require skilled clinicians and that algorithms need to be further optimized to manage complex PD symptoms, consensus was reached on aDBS safety and its ability to provide a faster and more stable treatment response than cDBS, also for tremor-dominant Parkinson's disease patients and for those with motor fluctuations and dyskinesias. Conclusions and Relevance Despite the need of further research, the panel concluded that aDBS is safe, promises to be maximally effective in PD patients with motor fluctuation and dyskinesias and therefore will enter into the clinical practice in the next years, with further research focused on algorithms and markers for complex symptoms.
Collapse
Affiliation(s)
- M. Guidetti
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - T. Bocci
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- Clinical Neurology Unit, “Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo”, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | | | - G. Deuschl
- Department of Neurology University Hospital Schleswig-Holstein, Campus Kiel and Christian Albrechts-University of Kiel Kiel Germany
| | - A. Fasano
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- CRANIA Center for Advancing Neurotechnological Innovation to Application, University of Toronto, ON, Canada
- KITE, University Health Network, Toronto, ON, Canada
- Edmond J. Safra Program in Parkinson’s Disease Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - R. Martinez Fernandez
- HM CINAC, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto Carlos III, CIBERNED, Madrid, Spain
| | - C. Gasca-Salas
- HM CINAC, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto Carlos III, CIBERNED, Madrid, Spain
| | - C. Hamani
- Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, M4N 3M5, ON, Canada
- Harquail Centre for Neuromodulation, 2075 Bayview Avenue, Toronto, M4N 3M5, ON, Canada
- Department of Surgery, University of Toronto, 149 College Street, Toronto, M5T 1P5, ON, Canada
| | - J.K. Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - A. A. Kühn
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität, Berlin, Germany
- NeuroCure, Exzellenzcluster, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZNE, German Center for Neurodegenerative Diseases, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| | - P. Limousin
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - S. Little
- Movement Disorders and Neuromodulation Centre, University of California San Francisco, San Francisco, California, USA
| | - A.M. Lozano
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- CRANIA Center for Advancing Neurotechnological Innovation to Application, University of Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - N.V. Maiorana
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - S. Marceglia
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - M.S. Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida, United States
- Department of Neurosurgery, Norman Fixel Institute for Neurological Diseases, University of Florida, United States
| | - S. Oliveri
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- Clinical Neurology Unit, “Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo”, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - J. L. Ostrem
- Movement Disorders and Neuromodulation Centre, University of California San Francisco, San Francisco, California, USA
| | - E. Scelzo
- Clinical Neurology Unit, “Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo”, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - A. Schnitzler
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Department of Neurology, Center for Movement Disorders and Neuromodulation, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - P.A. Starr
- UCSF Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- UCSF Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Y. Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - L. Timmermann
- Department of Neurology, University Hospital of Marburg, Marburg, Germany
| | - G. Tinkhauser
- Department of Neurology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - V. Visser-Vandewalle
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - J. Volkmann
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - A. Priori
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
- Clinical Neurology Unit, “Azienda Socio-Sanitaria Territoriale Santi Paolo e Carlo”, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| |
Collapse
|
13
|
Chung RS, Martin del Campo Vera R, Sundaram S, Cavaleri J, Gilbert ZD, Leonor A, Shao X, Zhang S, Kammen A, Mason X, Heck C, Liu CY, Kellis SS, Lee B. Beta-band power modulation in the human amygdala differentiates between go/no-go responses in an arm-reaching task. J Neural Eng 2024; 21:046019. [PMID: 38959877 PMCID: PMC11369913 DOI: 10.1088/1741-2552/ad5ebe] [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: 11/30/2023] [Revised: 04/22/2024] [Accepted: 07/03/2024] [Indexed: 07/05/2024]
Abstract
Objective. Traditionally known for its involvement in emotional processing, the amygdala's involvement in motor control remains relatively unexplored, with sparse investigations into the neural mechanisms governing amygdaloid motor movement and inhibition. This study aimed to characterize the amygdaloid beta-band (13-30 Hz) power between 'Go' and 'No-go' trials of an arm-reaching task.Approach. Ten participants with drug-resistant epilepsy implanted with stereoelectroencephalographic (SEEG) electrodes in the amygdala were enrolled in this study. SEEG data was recorded throughout discrete phases of a direct reach Go/No-go task, during which participants reached a touchscreen monitor or withheld movement based on a colored cue. Multitaper power analysis along with Wilcoxon signed-rank and Yates-correctedZtests were used to assess significant modulations of beta power between the Response and fixation (baseline) phases in the 'Go' and 'No-go' conditions.Main results. In the 'Go' condition, nine out of the ten participants showed a significant decrease in relative beta-band power during the Response phase (p⩽ 0.0499). In the 'No-go' condition, eight out of the ten participants presented a statistically significant increase in relative beta-band power during the response phase (p⩽ 0.0494). Four out of the eight participants with electrodes in the contralateral hemisphere and seven out of the eight participants with electrodes in the ipsilateral hemisphere presented significant modulation in beta-band power in both the 'Go' and 'No-go' conditions. At the group level, no significant differences were found between the contralateral and ipsilateral sides or between genders.Significance.This study reports beta-band power modulation in the human amygdala during voluntary movement in the setting of motor execution and inhibition. This finding supplements prior research in various brain regions associating beta-band power with motor control. The distinct beta-power modulation observed between these response conditions suggests involvement of amygdaloid oscillations in differentiating between motor inhibition and execution.
Collapse
Affiliation(s)
- Ryan S Chung
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Roberto Martin del Campo Vera
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Shivani Sundaram
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Jonathon Cavaleri
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Zachary D Gilbert
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Andrea Leonor
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Xiecheng Shao
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States of America
| | - Selena Zhang
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States of America
| | - Alexandra Kammen
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Xenos Mason
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States of America
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Christi Heck
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States of America
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Charles Y Liu
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States of America
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States of America
| | - Spencer S Kellis
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States of America
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
| | - Brian Lee
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States of America
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States of America
- Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States of America
| |
Collapse
|
14
|
Molina Galindo LS, Gonzalez-Escamilla G, Fleischer V, Grotegerd D, Meinert S, Ciolac D, Person M, Stein F, Brosch K, Nenadić I, Alexander N, Kircher T, Hahn T, Winter Y, Othman AE, Bittner S, Zipp F, Dannlowski U, Groppa S. Concurrent inflammation-related brain reorganization in multiple sclerosis and depression. Brain Behav Immun 2024; 119:978-988. [PMID: 38761819 DOI: 10.1016/j.bbi.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/02/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND Neuroinflammation affects brain tissue integrity in multiple sclerosis (MS) and may have a role in major depressive disorder (MDD). Whether advanced magnetic resonance imaging characteristics of the gray-to-white matter border serve as proxy of neuroinflammatory activity in MDD and MS remain unknown. METHODS We included 684 participants (132 MDD patients with recurrent depressive episodes (RDE), 70 MDD patients with a single depressive episode (SDE), 222 MS patients without depressive symptoms (nMS), 58 MS patients with depressive symptoms (dMS), and 202 healthy controls (HC)). 3 T-T1w MRI-derived gray-to-white matter contrast (GWc) was used to reconstruct and characterize connectivity alterations of GWc-covariance networks by means of modularity, clustering coefficient, and degree. A cross-validated support vector machine was used to test the ability of GWc to stratify groups according to their depression symptoms, measured with BDI, at the single-subject level in MS and MDD independently. FINDINGS MS and MDD patients showed increased modularity (ANOVA partial-η2 = 0.3) and clustering (partial-η2 = 0.1) compared to HC. In the subgroups, a linear trend analysis attested a gradient of modularity increases in the form: HC, dMS, nMS, SDE, and RDE (ANOVA partial-η2 = 0.28, p < 0.001) while this trend was less evident for clustering coefficient. Reduced morphological integrity (GWc) was seen in patients with increased depressive symptoms (partial-η2 = 0.42, P < 0.001) and was associated with depression scores across patient groups (r = -0.2, P < 0.001). Depressive symptoms in MS were robustly classified (88 %). CONCLUSIONS Similar structural network alterations in MDD and MS exist, suggesting possible common inflammatory events like demyelination, neuroinflammation that are caught by GWc analyses. These alterations may vary depending on the severity of symptoms and in the case of MS may elucidate the occurrence of comorbid depression.
Collapse
Affiliation(s)
- Lara S Molina Galindo
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Vinzenz Fleischer
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dumitru Ciolac
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Maren Person
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Frederike Stein
- Klinik für Psychiatrie und Psychotherapie, Philipps-Universität Marburg, Marburg, Germany
| | - Katharina Brosch
- Klinik für Psychiatrie und Psychotherapie, Philipps-Universität Marburg, Marburg, Germany
| | - Igor Nenadić
- Klinik für Psychiatrie und Psychotherapie, Philipps-Universität Marburg, Marburg, Germany
| | - Nina Alexander
- Klinik für Psychiatrie und Psychotherapie, Philipps-Universität Marburg, Marburg, Germany
| | - Tilo Kircher
- Klinik für Psychiatrie und Psychotherapie, Philipps-Universität Marburg, Marburg, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Yaroslav Winter
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Ahmed E Othman
- Department of Neuroradiology, Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Sergiu Groppa
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine-Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany.
| |
Collapse
|
15
|
Bange M, Gonzalez-Escamilla G, Herz DM, Tinkhauser G, Glaser M, Ciolac D, Pogosyan A, Kreis SL, Luhmann HJ, Tan H, Groppa S. Subthalamic stimulation modulates context-dependent effects of beta bursts during fine motor control. Nat Commun 2024; 15:3166. [PMID: 38605062 PMCID: PMC11009405 DOI: 10.1038/s41467-024-47555-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
Increasing evidence suggests a considerable role of pre-movement beta bursts for motor control and its impairment in Parkinson's disease. However, whether beta bursts occur during precise and prolonged movements and if they affect fine motor control remains unclear. To investigate the role of within-movement beta bursts for fine motor control, we here combine invasive electrophysiological recordings and clinical deep brain stimulation in the subthalamic nucleus in 19 patients with Parkinson's disease performing a context-varying task that comprised template-guided and free spiral drawing. We determined beta bursts in narrow frequency bands around patient-specific peaks and assessed burst amplitude, duration, and their immediate impact on drawing speed. We reveal that beta bursts occur during the execution of drawing movements with reduced duration and amplitude in comparison to rest. Exclusively when drawing freely, they parallel reductions in acceleration. Deep brain stimulation increases the acceleration around beta bursts in addition to a general increase in drawing velocity and improvements of clinical function. These results provide evidence for a diverse and task-specific role of subthalamic beta bursts for fine motor control in Parkinson's disease; suggesting that pathological beta bursts act in a context dependent manner, which can be targeted by clinical deep brain stimulation.
Collapse
Affiliation(s)
- Manuel Bange
- Section of Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Section of Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Damian M Herz
- Section of Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gerd Tinkhauser
- Department of Neurology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Martin Glaser
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dumitru Ciolac
- Section of Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Alek Pogosyan
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Svenja L Kreis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Huiling Tan
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sergiu Groppa
- Section of Movement Disorders and Neurostimulation, Department of Neurology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
16
|
Tian Y, Saradhi S, Bello E, Johnson MD, D’Eleuterio G, Popovic MR, Lankarany M. Model-based closed-loop control of thalamic deep brain stimulation. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1356653. [PMID: 38650608 PMCID: PMC11033853 DOI: 10.3389/fnetp.2024.1356653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
Introduction: Closed-loop control of deep brain stimulation (DBS) is beneficial for effective and automatic treatment of various neurological disorders like Parkinson's disease (PD) and essential tremor (ET). Manual (open-loop) DBS programming solely based on clinical observations relies on neurologists' expertise and patients' experience. Continuous stimulation in open-loop DBS may decrease battery life and cause side effects. On the contrary, a closed-loop DBS system uses a feedback biomarker/signal to track worsening (or improving) of patients' symptoms and offers several advantages compared to the open-loop DBS system. Existing closed-loop DBS control systems do not incorporate physiological mechanisms underlying DBS or symptoms, e.g., how DBS modulates dynamics of synaptic plasticity. Methods: In this work, we propose a computational framework for development of a model-based DBS controller where a neural model can describe the relationship between DBS and neural activity and a polynomial-based approximation can estimate the relationship between neural and behavioral activities. A controller is used in our model in a quasi-real-time manner to find DBS patterns that significantly reduce the worsening of symptoms. By using the proposed computational framework, these DBS patterns can be tested clinically by predicting the effect of DBS before delivering it to the patient. We applied this framework to the problem of finding optimal DBS frequencies for essential tremor given electromyography (EMG) recordings solely. Building on our recent network model of ventral intermediate nuclei (Vim), the main surgical target of the tremor, in response to DBS, we developed neural model simulation in which physiological mechanisms underlying Vim-DBS are linked to symptomatic changes in EMG signals. By using a proportional-integral-derivative (PID) controller, we showed that a closed-loop system can track EMG signals and adjust the stimulation frequency of Vim-DBS so that the power of EMG reaches a desired control target. Results and discussion: We demonstrated that the model-based DBS frequency aligns well with that used in clinical studies. Our model-based closed-loop system is adaptable to different control targets and can potentially be used for different diseases and personalized systems.
Collapse
Affiliation(s)
- Yupeng Tian
- Krembil Brain Institute—University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada
| | - Srikar Saradhi
- Krembil Brain Institute—University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Edward Bello
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Matthew D. Johnson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | | | - Milos R. Popovic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Milad Lankarany
- Krembil Brain Institute—University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application, University Health Network and University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Chao-Chia Lu D, Boulay C, Chan ADC, Sachs AJ. A Systematic Review of Neurophysiology-Based Localization Techniques Used in Deep Brain Stimulation Surgery of the Subthalamic Nucleus. Neuromodulation 2024; 27:409-421. [PMID: 37462595 DOI: 10.1016/j.neurom.2023.02.081] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 04/05/2024]
Abstract
OBJECTIVE This systematic review is conducted to identify, compare, and analyze neurophysiological feature selection, extraction, and classification to provide a comprehensive reference on neurophysiology-based subthalamic nucleus (STN) localization. MATERIALS AND METHODS The review was carried out using the methods and guidelines of the Kitchenham systematic review and provides an in-depth analysis on methods proposed on STN localization discussed in the literature between 2000 and 2021. Three research questions were formulated, and 115 publications were identified to answer the questions. RESULTS The three research questions formulated are answered using the literature found on the respective topics. This review discussed the technologies used in past research, and the performance of the state-of-the-art techniques is also reviewed. CONCLUSION This systematic review provides a comprehensive reference on neurophysiology-based STN localization by reviewing the research questions other new researchers may also have.
Collapse
Affiliation(s)
| | | | | | - Adam J Sachs
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
18
|
Abdulbaki A, Doll T, Helgers S, Heissler HE, Voges J, Krauss JK, Schwabe K, Alam M. Subthalamic Nucleus Deep Brain Stimulation Restores Motor and Sensorimotor Cortical Neuronal Oscillatory Activity in the Free-Moving 6-Hydroxydopamine Lesion Rat Parkinson Model. Neuromodulation 2024; 27:489-499. [PMID: 37002052 DOI: 10.1016/j.neurom.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVES Enhanced beta oscillations in cortical-basal ganglia (BG) thalamic circuitries have been linked to clinical symptoms of Parkinson's disease. Deep brain stimulation (DBS) of the subthalamic nucleus (STN) reduces beta band activity in BG regions, whereas little is known about activity in cortical regions. In this study, we investigated the effect of STN DBS on the spectral power of oscillatory activity in the motor cortex (MCtx) and sensorimotor cortex (SMCtx) by recording via an electrocorticogram (ECoG) array in free-moving 6-hydroxydopamine (6-OHDA) lesioned rats and sham-lesioned controls. MATERIALS AND METHODS Male Sprague-Dawley rats (250-350 g) were injected either with 6-OHDA or with saline in the right medial forebrain bundle, under general anesthesia. A stimulation electrode was then implanted in the ipsilateral STN, and an ECoG array was placed subdurally above the MCtx and SMCtx areas. Six days after the second surgery, the free-moving rats were individually recorded in three conditions: 1) basal activity, 2) during STN DBS, and 3) directly after STN DBS. RESULTS In 6-OHDA-lesioned rats (N = 8), the relative power of theta band activity was reduced, whereas activity of broad-range beta band (12-30 Hz) along with two different subbeta bands, that is, low (12-30 Hz) and high (20-30 Hz) beta band and gamma band, was higher in MCtx and SMCtx than in sham-lesioned controls (N = 7). This was, to some extent, reverted toward control level by STN DBS during and after stimulation. No major differences were found between contacts of the electrode grid or between MCtx and SMCtx. CONCLUSION Loss of nigrostriatal dopamine leads to abnormal oscillatory activity in both MCtx and SMCtx, which is compensated by STN stimulation, suggesting that parkinsonism-related oscillations in the cortex and BG are linked through their anatomic connections.
Collapse
Affiliation(s)
- Arif Abdulbaki
- Hannover Medical School, Department of Neurosurgery, Hannover, Germany.
| | - Theodor Doll
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Simeon Helgers
- Hannover Medical School, Department of Neurosurgery, Hannover, Germany
| | - Hans E Heissler
- Hannover Medical School, Department of Neurosurgery, Hannover, Germany
| | - Jürgen Voges
- Department of Stereotactic Neurosurgery, University Hospital Magdeburg, Magdeburg, Germany
| | - Joachim K Krauss
- Hannover Medical School, Department of Neurosurgery, Hannover, Germany
| | - Kerstin Schwabe
- Hannover Medical School, Department of Neurosurgery, Hannover, Germany
| | - Mesbah Alam
- Hannover Medical School, Department of Neurosurgery, Hannover, Germany
| |
Collapse
|
19
|
Carè M, Chiappalone M, Cota VR. Personalized strategies of neurostimulation: from static biomarkers to dynamic closed-loop assessment of neural function. Front Neurosci 2024; 18:1363128. [PMID: 38516316 PMCID: PMC10954825 DOI: 10.3389/fnins.2024.1363128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Despite considerable advancement of first choice treatment (pharmacological, physical therapy, etc.) over many decades, neurological disorders still represent a major portion of the worldwide disease burden. Particularly concerning, the trend is that this scenario will worsen given an ever expanding and aging population. The many different methods of brain stimulation (electrical, magnetic, etc.) are, on the other hand, one of the most promising alternatives to mitigate the suffering of patients and families when conventional treatment fall short of delivering efficacious treatment. With applications in virtually all neurological conditions, neurostimulation has seen considerable success in providing relief of symptoms. On the other hand, a large variability of therapeutic outcomes has also been observed, particularly in the usage of non-invasive brain stimulation (NIBS) modalities. Borrowing inspiration and concepts from its pharmacological counterpart and empowered by unprecedented neurotechnological advancement, the neurostimulation field has seen in recent years a widespread of methods aimed at the personalization of its parameters, based on biomarkers of the individuals being treated. The rationale is that, by taking into account important factors influencing the outcome, personalized stimulation can yield a much-improved therapy. Here, we review the literature to delineate the state-of-the-art of personalized stimulation, while also considering the important aspects of the type of informing parameter (anatomy, function, hybrid), invasiveness, and level of development (pre-clinical experimentation versus clinical trials). Moreover, by reviewing relevant literature on closed loop neuroengineering solutions in general and on activity dependent stimulation method in particular, we put forward the idea that improved personalization may be achieved when the method is able to track in real time brain dynamics and adjust its stimulation parameters accordingly. We conclude that such approaches have great potential of promoting the recovery of lost functions and enhance the quality of life for patients.
Collapse
Affiliation(s)
- Marta Carè
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Michela Chiappalone
- Department of Informatics, Bioengineering, Robotics System Engineering (DIBRIS), University of Genova, Genova, Italy
- Rehab Technologies Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | | |
Collapse
|
20
|
Anjum MF, Espinoza AI, Cole RC, Singh A, May P, Uc EY, Dasgupta S, Narayanan NS. Resting-state EEG measures cognitive impairment in Parkinson's disease. NPJ Parkinsons Dis 2024; 10:6. [PMID: 38172519 PMCID: PMC10764756 DOI: 10.1038/s41531-023-00602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
Cognitive dysfunction is common in Parkinson's disease (PD). We developed and evaluated an EEG-based biomarker to index cognitive functions in PD from a few minutes of resting-state EEG. We hypothesized that synchronous changes in EEG across the power spectrum can measure cognition. We optimized a data-driven algorithm to efficiently capture these changes and index cognitive function in 100 PD and 49 control participants. We compared our EEG-based cognitive index with the Montreal cognitive assessment (MoCA) and cognitive tests across different domains from National Institutes of Health (NIH) Toolbox using cross-validations, regression models, and randomization tests. Finally, we externally validated our approach on 32 PD participants. We observed cognition-related changes in EEG over multiple spectral rhythms. Utilizing only 8 best-performing electrodes, our proposed index strongly correlated with cognition (MoCA: rho = 0.68, p value < 0.001; NIH-Toolbox cognitive tests: rho ≥ 0.56, p value < 0.001) outperforming traditional spectral markers (rho = -0.30-0.37). The index showed a strong fit in regression models (R2 = 0.46) with MoCA, yielded 80% accuracy in detecting cognitive impairment, and was effective in both PD and control participants. Notably, our approach was equally effective (rho = 0.68, p value < 0.001; MoCA) in out-of-sample testing. In summary, we introduced a computationally efficient data-driven approach for cross-domain cognition indexing using fewer than 10 EEG electrodes, potentially compatible with dynamic therapies like closed-loop neurostimulation. These results will inform next-generation neurophysiological biomarkers for monitoring cognition in PD and other neurological diseases.
Collapse
Affiliation(s)
- Md Fahim Anjum
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Arturo I Espinoza
- Department of Neurology, The University of Iowa, Iowa city, IA, 52240, USA
| | - Rachel C Cole
- Department of Neurology, The University of Iowa, Iowa city, IA, 52240, USA
| | - Arun Singh
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, South Dakota, SD, 57069, USA
| | - Patrick May
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa city, IA, 52240, USA
| | - Ergun Y Uc
- Department of Neurology, The University of Iowa, Iowa city, IA, 52240, USA
- Neurology Service, Iowa City VA Medical Center, Iowa city, IA, 52240, USA
| | - Soura Dasgupta
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa city, IA, 52240, USA
| | | |
Collapse
|
21
|
Krishnan J, Joseph R, Vayalappil MC, Krishnan S, Kishore A. A Review on Implantable Neuroelectrodes. Crit Rev Biomed Eng 2024; 52:21-39. [PMID: 37938182 DOI: 10.1615/critrevbiomedeng.2023049282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
The efficacy of every neuromodulation modality depends upon the characteristics of the electrodes used to stimulate the chosen target. The geometrical, chemical, mechanical and physical configuration of electrodes used in neurostimulation affects several performance attributes like stimulation efficiency, selectivity, tissue response, etc. The efficiency of stimulation in relation to electrode impedance is influenced by the electrode material and/or its geometry. The nature of the electrode material determines the charge transfer across the electrode-tissue interface, which also relates to neuronal tissue damage. Electrode morphology or configuration pattern can facilitate the modulation of extracellular electric field (field shaping). This enables selective activation of neurons and minimizes side effects. Biocompatibility and biostability of the electrode materials or electrode coating have a role in glial formation and tissue damage. Mechanical and electrochemical stability (corrosion resistance) determines the long-term efficacy of any neuromodulation technique. Here, a review of electrodes typically used for implantable neuromodulation is discussed. Factors affecting the performance of electrodes like stimulation efficiency, selectivity and tissue responses to the electrode-tissue interface are discussed. Technological advancements to improve electrode characteristics are also included.
Collapse
Affiliation(s)
- Jithin Krishnan
- Department of Medical Devices Engineering, BMT Wing, SCTIMST, Kerala, India
| | - Roy Joseph
- Department of Medical Devices Engineering, BMT Wing, SCTIMST, Kerala, India
| | | | | | - Asha Kishore
- Aster Parkinson & Movement Disorder Centre, Senior Consultant Neurologist and Movement Disorder Specialist
| |
Collapse
|
22
|
Hill ME, Johnson LA, Wang J, Sanabria DE, Patriat R, Cooper SE, Park MC, Harel N, Vitek JL, Aman JE. Paradoxical Modulation of STN β-Band Activity with Medication Compared to Deep Brain Stimulation. Mov Disord 2024; 39:192-197. [PMID: 37888906 PMCID: PMC10843006 DOI: 10.1002/mds.29634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Excessive subthalamic nucleus (STN) β-band (13-35 Hz) synchronized oscillations has garnered interest as a biomarker for characterizing disease state and developing adaptive stimulation systems for Parkinson's disease (PD). OBJECTIVES To report on a patient with abnormal treatment-responsive modulation in the β-band. METHODS We examined STN local field potentials from an externalized deep brain stimulation (DBS) lead while assessing PD motor signs in four conditions (OFF, MEDS, DBS, and MEDS+DBS). RESULTS The patient presented here exhibited a paradoxical increase in β power following administration of levodopa and pramipexole (MEDS), but an attenuation in β power during DBS and MEDS+DBS despite clinical improvement of 50% or greater under all three therapeutic conditions. CONCLUSIONS This case highlights the need for further study on the role of β oscillations in the pathophysiology of PD and the importance of personalized approaches to the development of β or other biomarker-based DBS closed loop algorithms. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Meghan E. Hill
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Luke A. Johnson
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Jing Wang
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | | | - Rémi Patriat
- Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Scott E. Cooper
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Michael C. Park
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Noam Harel
- Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Jerrold L. Vitek
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Joshua E. Aman
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
23
|
He S, Baig F, Merla A, Torrecillos F, Perera A, Wiest C, Debarros J, Benjaber M, Hart MG, Ricciardi L, Morgante F, Hasegawa H, Samuel M, Edwards M, Denison T, Pogosyan A, Ashkan K, Pereira E, Tan H. Beta-triggered adaptive deep brain stimulation during reaching movement in Parkinson's disease. Brain 2023; 146:5015-5030. [PMID: 37433037 PMCID: PMC10690014 DOI: 10.1093/brain/awad233] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/30/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Subthalamic nucleus (STN) beta-triggered adaptive deep brain stimulation (ADBS) has been shown to provide clinical improvement comparable to conventional continuous DBS (CDBS) with less energy delivered to the brain and less stimulation induced side effects. However, several questions remain unanswered. First, there is a normal physiological reduction of STN beta band power just prior to and during voluntary movement. ADBS systems will therefore reduce or cease stimulation during movement in people with Parkinson's disease and could therefore compromise motor performance compared to CDBS. Second, beta power was smoothed and estimated over a time period of 400 ms in most previous ADBS studies, but a shorter smoothing period could have the advantage of being more sensitive to changes in beta power, which could enhance motor performance. In this study, we addressed these two questions by evaluating the effectiveness of STN beta-triggered ADBS using a standard 400 ms and a shorter 200 ms smoothing window during reaching movements. Results from 13 people with Parkinson's disease showed that reducing the smoothing window for quantifying beta did lead to shortened beta burst durations by increasing the number of beta bursts shorter than 200 ms and more frequent switching on/off of the stimulator but had no behavioural effects. Both ADBS and CDBS improved motor performance to an equivalent extent compared to no DBS. Secondary analysis revealed that there were independent effects of a decrease in beta power and an increase in gamma power in predicting faster movement speed, while a decrease in beta event related desynchronization (ERD) predicted quicker movement initiation. CDBS suppressed both beta and gamma more than ADBS, whereas beta ERD was reduced to a similar level during CDBS and ADBS compared with no DBS, which together explained the achieved similar performance improvement in reaching movements during CDBS and ADBS. In addition, ADBS significantly improved tremor compared with no DBS but was not as effective as CDBS. These results suggest that STN beta-triggered ADBS is effective in improving motor performance during reaching movements in people with Parkinson's disease, and that shortening of the smoothing window does not result in any additional behavioural benefit. When developing ADBS systems for Parkinson's disease, it might not be necessary to track very fast beta dynamics; combining beta, gamma, and information from motor decoding might be more beneficial with additional biomarkers needed for optimal treatment of tremor.
Collapse
Affiliation(s)
- Shenghong He
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Fahd Baig
- Neurosciences Research Centre, St George’s, University of London & St George’s University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Cranmer Terrace, London SW17 0QT, UK
| | - Anca Merla
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Flavie Torrecillos
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Andrea Perera
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Christoph Wiest
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Jean Debarros
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Moaad Benjaber
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Michael G Hart
- Neurosciences Research Centre, St George’s, University of London & St George’s University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Cranmer Terrace, London SW17 0QT, UK
| | - Lucia Ricciardi
- Neurosciences Research Centre, St George’s, University of London & St George’s University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Cranmer Terrace, London SW17 0QT, UK
| | - Francesca Morgante
- Neurosciences Research Centre, St George’s, University of London & St George’s University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Cranmer Terrace, London SW17 0QT, UK
| | - Harutomo Hasegawa
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Michael Samuel
- Department of Neurology, King’s College Hospital NHS Foundation Trust, London, SE5 9RS, UK
| | - Mark Edwards
- Department of Clinical and Basic Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London WC2R 2LS, UK
| | - Timothy Denison
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Alek Pogosyan
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Keyoumars Ashkan
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Erlick Pereira
- Neurosciences Research Centre, St George’s, University of London & St George’s University Hospitals NHS Foundation Trust, Institute of Molecular and Clinical Sciences, Cranmer Terrace, London SW17 0QT, UK
| | - Huiling Tan
- MRC Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
24
|
Xu W, Wang J, Li XN, Liang J, Song L, Wu Y, Liu Z, Sun B, Li WG. Neuronal and synaptic adaptations underlying the benefits of deep brain stimulation for Parkinson's disease. Transl Neurodegener 2023; 12:55. [PMID: 38037124 PMCID: PMC10688037 DOI: 10.1186/s40035-023-00390-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: 08/01/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
Deep brain stimulation (DBS) is a well-established and effective treatment for patients with advanced Parkinson's disease (PD), yet its underlying mechanisms remain enigmatic. Optogenetics, primarily conducted in animal models, provides a unique approach that allows cell type- and projection-specific modulation that mirrors the frequency-dependent stimulus effects of DBS. Opto-DBS research in animal models plays a pivotal role in unraveling the neuronal and synaptic adaptations that contribute to the efficacy of DBS in PD treatment. DBS-induced neuronal responses rely on a complex interplay between the distributions of presynaptic inputs, frequency-dependent synaptic depression, and the intrinsic excitability of postsynaptic neurons. This orchestration leads to conversion of firing patterns, enabling both antidromic and orthodromic modulation of neural circuits. Understanding these mechanisms is vital for decoding position- and programming-dependent effects of DBS. Furthermore, patterned stimulation is emerging as a promising strategy yielding long-lasting therapeutic benefits. Research on the neuronal and synaptic adaptations to DBS may pave the way for the development of more enduring and precise modulation patterns. Advanced technologies, such as adaptive DBS or directional electrodes, can also be integrated for circuit-specific neuromodulation. These insights hold the potential to greatly improve the effectiveness of DBS and advance PD treatment to new levels.
Collapse
Affiliation(s)
- Wenying Xu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xin-Ni Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jingxue Liang
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- 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
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
- Ministry of Education-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
25
|
Oliveira AM, Coelho L, Carvalho E, Ferreira-Pinto MJ, Vaz R, Aguiar P. Machine learning for adaptive deep brain stimulation in Parkinson's disease: closing the loop. J Neurol 2023; 270:5313-5326. [PMID: 37530789 PMCID: PMC10576725 DOI: 10.1007/s00415-023-11873-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease bearing a severe social and economic impact. So far, there is no known disease modifying therapy and the current available treatments are symptom oriented. Deep Brain Stimulation (DBS) is established as an effective treatment for PD, however current systems lag behind today's technological potential. Adaptive DBS, where stimulation parameters depend on the patient's physiological state, emerges as an important step towards "smart" DBS, a strategy that enables adaptive stimulation and personalized therapy. This new strategy is facilitated by currently available neurotechnologies allowing the simultaneous monitoring of multiple signals, providing relevant physiological information. Advanced computational models and analytical methods are an important tool to explore the richness of the available data and identify signal properties to close the loop in DBS. To tackle this challenge, machine learning (ML) methods applied to DBS have gained popularity due to their ability to make good predictions in the presence of multiple variables and subtle patterns. ML based approaches are being explored at different fronts such as the identification of electrophysiological biomarkers and the development of personalized control systems, leading to effective symptom relief. In this review, we explore how ML can help overcome the challenges in the development of closed-loop DBS, particularly its role in the search for effective electrophysiology biomarkers. Promising results demonstrate ML potential for supporting a new generation of adaptive DBS, with better management of stimulation delivery, resulting in more efficient and patient-tailored treatments.
Collapse
Affiliation(s)
- Andreia M Oliveira
- Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
- Neuroengineering and Computational Neuroscience Lab, Instituto de Investigação e Inovação da Universidade do Porto, Porto, Portugal
| | - Luis Coelho
- Instituto Superior de Engenharia do Porto, Porto, Portugal
| | - Eduardo Carvalho
- Neuroengineering and Computational Neuroscience Lab, Instituto de Investigação e Inovação da Universidade do Porto, Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Manuel J Ferreira-Pinto
- Centro Hospitalar Universitário de São João, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Rui Vaz
- Centro Hospitalar Universitário de São João, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Paulo Aguiar
- Faculdade de Engenharia da Universidade do Porto, Porto, Portugal.
- Neuroengineering and Computational Neuroscience Lab, Instituto de Investigação e Inovação da Universidade do Porto, Porto, Portugal.
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
| |
Collapse
|
26
|
Radcliffe EM, Baumgartner AJ, Kern DS, Al Borno M, Ojemann S, Kramer DR, Thompson JA. Oscillatory beta dynamics inform biomarker-driven treatment optimization for Parkinson's disease. J Neurophysiol 2023; 129:1492-1504. [PMID: 37198135 DOI: 10.1152/jn.00055.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/23/2023] [Accepted: 05/17/2023] [Indexed: 05/19/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by loss of dopaminergic neurons and dysregulation of the basal ganglia. Cardinal motor symptoms include bradykinesia, rigidity, and tremor. Deep brain stimulation (DBS) of select subcortical nuclei is standard of care for medication-refractory PD. Conventional open-loop DBS delivers continuous stimulation with fixed parameters that do not account for a patient's dynamic activity state or medication cycle. In comparison, closed-loop DBS, or adaptive DBS (aDBS), adjusts stimulation based on biomarker feedback that correlates with clinical state. Recent work has identified several neurophysiological biomarkers in local field potential recordings from PD patients, the most promising of which are 1) elevated beta (∼13-30 Hz) power in the subthalamic nucleus (STN), 2) increased beta synchrony throughout basal ganglia-thalamocortical circuits, notably observed as coupling between the STN beta phase and cortical broadband gamma (∼50-200 Hz) amplitude, and 3) prolonged beta bursts in the STN and cortex. In this review, we highlight relevant frequency and time domain features of STN beta measured in PD patients and summarize how spectral beta power, oscillatory beta synchrony, phase-amplitude coupling, and temporal beta bursting inform PD pathology, neurosurgical targeting, and DBS therapy. We then review how STN beta dynamics inform predictive, biomarker-driven aDBS approaches for optimizing PD treatment. We therefore provide clinically useful and actionable insight that can be applied toward aDBS implementation for PD.
Collapse
Affiliation(s)
- Erin M Radcliffe
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Alexander J Baumgartner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Drew S Kern
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Mazen Al Borno
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Computer Science and Engineering, University of Colorado Denver, Denver, Colorado, United States
| | - Steven Ojemann
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Daniel R Kramer
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - John A Thompson
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
27
|
Neumann WJ, Gilron R, Little S, Tinkhauser G. Adaptive Deep Brain Stimulation: From Experimental Evidence Toward Practical Implementation. Mov Disord 2023. [PMID: 37148553 DOI: 10.1002/mds.29415] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 03/27/2023] [Accepted: 04/05/2023] [Indexed: 05/08/2023] Open
Abstract
Closed-loop adaptive deep brain stimulation (aDBS) can deliver individualized therapy at an unprecedented temporal precision for neurological disorders. This has the potential to lead to a breakthrough in neurotechnology, but the translation to clinical practice remains a significant challenge. Via bidirectional implantable brain-computer-interfaces that have become commercially available, aDBS can now sense and selectively modulate pathophysiological brain circuit activity. Pilot studies investigating different aDBS control strategies showed promising results, but the short experimental study designs have not yet supported individualized analyses of patient-specific factors in biomarker and therapeutic response dynamics. Notwithstanding the clear theoretical advantages of a patient-tailored approach, these new stimulation possibilities open a vast and mostly unexplored parameter space, leading to practical hurdles in the implementation and development of clinical trials. Therefore, a thorough understanding of the neurophysiological and neurotechnological aspects related to aDBS is crucial to develop evidence-based treatment regimens for clinical practice. Therapeutic success of aDBS will depend on the integrated development of strategies for feedback signal identification, artifact mitigation, signal processing, and control policy adjustment, for precise stimulation delivery tailored to individual patients. The present review introduces the reader to the neurophysiological foundation of aDBS for Parkinson's disease (PD) and other network disorders, explains currently available aDBS control policies, and highlights practical pitfalls and difficulties to be addressed in the upcoming years. Finally, it highlights the importance of interdisciplinary clinical neurotechnological research within and across DBS centers, toward an individualized patient-centered approach to invasive brain stimulation. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Wolf-Julian Neumann
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Simon Little
- Movement Disorders and Neuromodulation Centre, University of California San Francisco, San Francisco, California, USA
| | - Gerd Tinkhauser
- Department of Neurology, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Widge AS. Closed-Loop Deep Brain Stimulation for Psychiatric Disorders. Harv Rev Psychiatry 2023; 31:162-171. [PMID: 37171475 PMCID: PMC10188203 DOI: 10.1097/hrp.0000000000000367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
ABSTRACT Deep brain stimulation (DBS) is a well-established approach to treating medication-refractory neurological disorders and holds promise for treating psychiatric disorders. Despite strong open-label results in extremely refractory patients, DBS has struggled to meet endpoints in randomized controlled trials. A major challenge is stimulation "dosing"-DBS systems have many adjustable parameters, and clinicians receive little feedback on whether they have chosen the correct parameters for an individual patient. Multiple groups have proposed closed loop technologies as a solution. These systems sense electrical activity, identify markers of an (un)desired state, then automatically deliver or adjust stimulation to alter that electrical state. Closed loop DBS has been successfully deployed in movement disorders and epilepsy. The availability of that technology, as well as advances in opportunities for invasive research with neurosurgical patients, has yielded multiple pilot demonstrations in psychiatric illness. Those demonstrations split into two schools of thought, one rooted in well-established diagnoses and symptom scales, the other in the more experimental Research Domain Criteria (RDoC) framework. Both are promising, and both are limited by the boundaries of current stimulation technology. They are in turn driving advances in implantable recording hardware, signal processing, and stimulation paradigms. The combination of these advances is likely to change both our understanding of psychiatric neurobiology and our treatment toolbox, though the timeframe may be limited by the realities of implantable device development.
Collapse
Affiliation(s)
- Alik S Widge
- From the Department of Psychiatry & Behavioral Sciences and Medical Discovery Team on Addictions, University of Minnesota
| |
Collapse
|
29
|
Kurtin DL, Giunchiglia V, Vohryzek J, Cabral J, Skeldon AC, Violante IR. Moving from phenomenological to predictive modelling: Progress and pitfalls of modelling brain stimulation in-silico. Neuroimage 2023; 272:120042. [PMID: 36965862 DOI: 10.1016/j.neuroimage.2023.120042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/06/2023] [Accepted: 03/16/2023] [Indexed: 03/27/2023] Open
Abstract
Brain stimulation is an increasingly popular neuromodulatory tool used in both clinical and research settings; however, the effects of brain stimulation, particularly those of non-invasive stimulation, are variable. This variability can be partially explained by an incomplete mechanistic understanding, coupled with a combinatorial explosion of possible stimulation parameters. Computational models constitute a useful tool to explore the vast sea of stimulation parameters and characterise their effects on brain activity. Yet the utility of modelling stimulation in-silico relies on its biophysical relevance, which needs to account for the dynamics of large and diverse neural populations and how underlying networks shape those collective dynamics. The large number of parameters to consider when constructing a model is no less than those needed to consider when planning empirical studies. This piece is centred on the application of phenomenological and biophysical models in non-invasive brain stimulation. We first introduce common forms of brain stimulation and computational models, and provide typical construction choices made when building phenomenological and biophysical models. Through the lens of four case studies, we provide an account of the questions these models can address, commonalities, and limitations across studies. We conclude by proposing future directions to fully realise the potential of computational models of brain stimulation for the design of personalized, efficient, and effective stimulation strategies.
Collapse
Affiliation(s)
- Danielle L Kurtin
- Neuromodulation Laboratory, School of Psychology, University of Surrey, Guildford, GU2 7XH, United Kingdom; Department of Brain Sciences, Imperial College London, London, United Kingdom.
| | | | - Jakub Vohryzek
- Centre for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain; Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, UK
| | - Joana Cabral
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Anne C Skeldon
- Department of Mathematics, Centre for Mathematical and Computational Biology, University of Surrey, Guildford, United Kingdom
| | - Ines R Violante
- Neuromodulation Laboratory, School of Psychology, University of Surrey, Guildford, GU2 7XH, United Kingdom
| |
Collapse
|
30
|
Anjum MF, Espinoza A, Cole R, Singh A, May P, Uc E, Dasgupta S, Narayanan N. Resting-state EEG measures cognitive impairment in Parkinson's disease. RESEARCH SQUARE 2023:rs.3.rs-2666578. [PMID: 36993450 PMCID: PMC10055637 DOI: 10.21203/rs.3.rs-2666578/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Background Cognitive dysfunction is common in Parkinson's disease (PD) and is diagnosed by complex, time-consuming psychometric tests which are affected by language and education, subject to learning effects, and not suitable for continuous monitoring of cognition. Objectives We developed and evaluated an EEG-based biomarker to index cognitive functions in PD from a few minutes of resting-state EEG. Methods We hypothesized that synchronous changes in EEG across the power spectrum can measure cognition. We optimized a data-driven algorithm to efficiently capture these changes and index cognitive function in 100 PD and 49 control participants. We compared our EEG-based cognitive index with the Montreal cognitive assessment (MoCA) and cognitive tests across different domains from the National Institutes of Health (NIH) Toolbox using cross-validation schemes, regression models, and randomization tests. Results We observed cognition-related changes in EEG activities over multiple spectral rhythms. Utilizing only 8 best-performing EEG electrodes, our proposed index strongly correlated with cognition (rho = 0.68, p value < 0.001 with MoCA; rho ≥ 0.56, p value < 0.001 with cognitive tests from the NIH Toolbox) outperforming traditional spectral markers (rho = -0.30 - 0.37). The index showed a strong fit in regression models (R2 = 0.46) with MoCA, yielded 80% accuracy in detecting cognitive impairment, and was effective in both PD and control participants. Conclusions Our approach is computationally efficient for real-time indexing of cognition across domains, implementable even in hardware with limited computing capabilities, making it potentially compatible with dynamic therapies such as closed-loop neurostimulation, and will inform next-generation neurophysiological biomarkers for monitoring cognition in PD and other neurological diseases.
Collapse
|
31
|
Sindhu KR, Ngo D, Ombao H, Olaya JE, Shrey DW, Lopour BA. A novel method for dynamically altering the surface area of intracranial EEG electrodes. J Neural Eng 2023; 20:026002. [PMID: 36720162 PMCID: PMC9990369 DOI: 10.1088/1741-2552/acb79f] [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: 01/05/2023] [Accepted: 01/31/2023] [Indexed: 02/02/2023]
Abstract
Objective.Intracranial electroencephalogram (iEEG) plays a critical role in the treatment of neurological diseases, such as epilepsy and Parkinson's disease, as well as the development of neural prostheses and brain computer interfaces. While electrode geometries vary widely across these applications, the impact of electrode size on iEEG features and morphology is not well understood. Some insight has been gained from computer simulations, as well as experiments in which signals are recorded using electrodes of different sizes concurrently in different brain regions. Here, we introduce a novel method to record from electrodes of different sizes in the exact same location by changing the size of iEEG electrodes after implantation in the brain.Approach.We first present a theoretical model and anin vitrovalidation of the method. We then report the results of anin vivoimplementation in three human subjects with refractory epilepsy. We recorded iEEG data from three different electrode sizes and compared the amplitudes, power spectra, inter-channel correlations, and signal-to-noise ratio (SNR) of interictal epileptiform discharges, i.e. epileptic spikes.Main Results.We found that iEEG amplitude and power decreased as electrode size increased, while inter-channel correlation did not change significantly with electrode size. The SNR of epileptic spikes was generally highest in the smallest electrodes, but 39% of spikes had maximal SNR in larger electrodes. This likely depends on the precise location and spatial spread of each spike.Significance.Overall, this new method enables multi-scale measurements of electrical activity in the human brain that can facilitate our understanding of neurophysiology, treatment of neurological disease, and development of novel technologies.
Collapse
Affiliation(s)
| | - Duy Ngo
- Department of Statistics, Western Michigan University, Kalamazoo, MI, United States of America
| | - Hernando Ombao
- Statistics Program, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Joffre E Olaya
- Division of Neurosurgery, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Neurosurgery, University of California, Irvine, Irvine, CA, United States of America
| | - Daniel W Shrey
- Division of Neurology, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States of America
| | - Beth A Lopour
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States of America
| |
Collapse
|
32
|
An Q, Yin Z, Ma R, Fan H, Xu Y, Gan Y, Gao Y, Meng F, Yang A, Jiang Y, Zhu G, Zhang J. Adaptive deep brain stimulation for Parkinson's disease: looking back at the past decade on motor outcomes. J Neurol 2023; 270:1371-1387. [PMID: 36471098 DOI: 10.1007/s00415-022-11495-z] [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: 07/30/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Adaptive deep brain stimulation (aDBS) has been reported to be an effective treatment for motor symptoms in patients with Parkinson's disease (PD). However, it remains unclear whether and in which motor domain aDBS provides greater/less benefits than conventional DBS (cDBS). OBJECTIVE To conduct a meta-analysis and systematic review to explore the improvement of the motor symptoms of PD patients undergoing aDBS and the comparison between aDBS and cDBS. METHODS Nineteen studies from PubMed, Embase, and the Cochrane Library database were eligible for the main analysis. Twelve studies used quantitative plus qualitative analysis; seven studies were only qualitatively analyzed. The efficacy of aDBS was evaluated and compared to cDBS through overall motor function improvements, changes in symptoms of rigidity-bradykinesia, dyskinesia, tremor, and speech function, and total electrical energy delivered (TEED). The overall motor improvement and TEED were investigated through meta-analyses, while other variables were investigated by systematic review. RESULTS Quantitative analysis showed that aDBS, with a reduction of TEED (55% of that of cDBS), significantly improved motor functions (33.9%, p < 0.01) and may be superior to cDBS in overall motor improvement (p = 0.002). However, significant publication bias was detected regarding the superiority (p = 0.006, Egger's test). In the qualitative analysis, rigidity-bradykinesia, dyskinesia, and speech function outcomes after aDBS and cDBS were comparable. Beta-based aDBS may not be as efficient as cDBS for tremor control. CONCLUSIONS aDBS can effectively relieve the clinical symptoms of advanced PD as did cDBS, at least in acute trials, delivering less stimulation than cDBS. Specific symptoms including tremor and axial disability remain to be compared between aDBS and cDBS in long-term studies.
Collapse
Affiliation(s)
- Qi An
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Zixiao Yin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Ruoyu Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Houyou Fan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Yichen Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Yifei Gan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Yuan Gao
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Fangang Meng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China.,Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Anchao Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Yin Jiang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China
| | - Guanyu Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China.
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China. .,Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, 100070, Beijing, China. .,Beijing Key Laboratory of Neurostimulation, Beijing, China.
| |
Collapse
|
33
|
Morelli N, Summers RLS. Association of subthalamic beta frequency sub-bands to symptom severity in patients with Parkinson's disease: A systematic review. Parkinsonism Relat Disord 2023; 110:105364. [PMID: 36997437 DOI: 10.1016/j.parkreldis.2023.105364] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
OBJECTIVE Local field potentials (LFP), specifically beta (13-30Hz) frequency measures, have been found to be associated with motor dysfunction in people with Parkinson's disease (PwPD). A consensus on beta subband (low- and high-beta) relationships to clinical state or therapy response has yet to be determined. The objective of this review is to synthesize literature reporting the association of low- and high-beta characteristics to clinical ratings of motor symptoms in PwPD. METHODS A systematic search of existing literature was completed using EMBASE. Articles which collected subthalamic nucleus (STN) LFPs using macroelectrodes in PwPD, analyzed low- (13-20 Hz) and high-beta (21-35 Hz) bands, collected UPDRS-III, and reported correlational strength or predictive capacity of LFPs to UPDRS-III scores. RESULTS The initial search yielded 234 articles, with 11 articles achieving inclusion. Beta measures included power spectral density, peak characteristics, and burst characteristics. High-beta was a significant predictor of UPDRS-III responses to therapy in 5 (100%) articles. Low-beta was significantly associated with UPDRS-III total score in 3 (60%) articles. Low- and high-beta associations to UPDRS-III subscores were mixed. CONCLUSION This systematic review reinforces previous reports that beta band oscillatory measures demonstrate a consistent relationship to Parkinsonian motor symptoms and ability to predict motor response to therapy. Specifically, high-beta, demonstrated a consistent ability to predict UPDRS-III responses to common PD therapies, while low-beta measures were associated with general Parkinsonian symptom severity. Continued research is needed to determine which beta subband demonstrates the greatest association to motor symptom subtypes and potentially offers clinical utility toward LFP-guided DBS programming and adaptive DBS.
Collapse
|
34
|
Bahadori-Jahromi F, Salehi S, Madadi Asl M, Valizadeh A. Efficient suppression of parkinsonian beta oscillations in a closed-loop model of deep brain stimulation with amplitude modulation. Front Hum Neurosci 2023; 16:1013155. [PMID: 36776221 PMCID: PMC9908610 DOI: 10.3389/fnhum.2022.1013155] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/09/2022] [Indexed: 01/27/2023] Open
Abstract
Introduction Parkinson's disease (PD) is a movement disorder characterized by the pathological beta band (15-30 Hz) neural oscillations within the basal ganglia (BG). It is shown that the suppression of abnormal beta oscillations is correlated with the improvement of PD motor symptoms, which is a goal of standard therapies including deep brain stimulation (DBS). To overcome the stimulation-induced side effects and inefficiencies of conventional DBS (cDBS) and to reduce the administered stimulation current, closed-loop adaptive DBS (aDBS) techniques were developed. In this method, the frequency and/or amplitude of stimulation are modulated based on various disease biomarkers. Methods Here, by computational modeling of a cortico-BG-thalamic network in normal and PD conditions, we show that closed-loop aDBS of the subthalamic nucleus (STN) with amplitude modulation leads to a more effective suppression of pathological beta oscillations within the parkinsonian BG. Results Our results show that beta band neural oscillations are restored to their normal range and the reliability of the response of the thalamic neurons to motor cortex commands is retained due to aDBS with amplitude modulation. Furthermore, notably less stimulation current is administered during aDBS compared with cDBS due to a closed-loop control of stimulation amplitude based on the STN local field potential (LFP) beta activity. Discussion Efficient models of closed-loop stimulation may contribute to the clinical development of optimized aDBS techniques designed to reduce potential stimulation-induced side effects of cDBS in PD patients while leading to a better therapeutic outcome.
Collapse
Affiliation(s)
| | - Sina Salehi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Madadi Asl
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran, Iran
| | - Alireza Valizadeh
- Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran, Iran
| |
Collapse
|
35
|
Association between Beta Oscillations from Subthalamic Nucleus and Quantitative Susceptibility Mapping in Deep Gray Matter Structures in Parkinson's Disease. Brain Sci 2023; 13:brainsci13010081. [PMID: 36672062 PMCID: PMC9857066 DOI: 10.3390/brainsci13010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/15/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
This study aimed to investigate the association between beta oscillations and brain iron deposition. Beta oscillations were filtered from the microelectrode recordings of local field potentials (LFP) in the subthalamic nucleus (STN), and the ratio of the power spectral density of beta oscillations (PSDXb) to that of the LFP signals was calculated. Iron deposition in the deep gray matter (DGM) structures was indirectly assessed using quantitative susceptibility mapping (QSM). The Unified Parkinson's Disease Rating Scale (UPDRS), part III, was used to assess the severity of symptoms. Spearman correlation coefficients were applied to assess the associations of PSDXb with QSM values in the DGM structures and the severity of symptoms. PSDXb showed a significant positive correlation with the average QSM values in DGM structures, including caudate and substantia nigra (SN) (p = 0.008 and 0.044). Similarly, the PSDXb showed significant negative correlations with the severity of symptoms, including axial symptoms and the gait in the medicine-off state (p = 0.006 for both). The abnormal iron metabolism in the SN and striatum pathways may be one of the underlying mechanisms for the occurrence of abnormal beta oscillations in the STN, and beta oscillations may serve as important pathophysiological biomarkers of PD.
Collapse
|
36
|
Peterson V, Merk T, Bush A, Nikulin V, Kühn AA, Neumann WJ, Richardson RM. Movement decoding using spatio-spectral features of cortical and subcortical local field potentials. Exp Neurol 2023; 359:114261. [PMID: 36349662 DOI: 10.1016/j.expneurol.2022.114261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 09/26/2022] [Accepted: 10/25/2022] [Indexed: 12/30/2022]
Abstract
The first commercially sensing enabled deep brain stimulation (DBS) devices for the treatment of movement disorders have recently become available. In the future, such devices could leverage machine learning based brain signal decoding strategies to individualize and adapt therapy in real-time. As multi-channel recordings become available, spatial information may provide an additional advantage for informing machine learning models. To investigate this concept, we compared decoding performances from single channels vs. spatial filtering techniques using intracerebral multitarget electrophysiology in Parkinson's disease patients undergoing DBS implantation. We investigated the feasibility of spatial filtering in invasive neurophysiology and the putative utility of combined cortical ECoG and subthalamic local field potential signals for decoding grip-force, a well-defined and continuous motor readout. We found that adding spatial information to the model can improve decoding (6% gain in decoding), but the spatial patterns and additional benefit was highly individual. Beyond decoding performance results, spatial filters and patterns can be used to obtain meaningful neurophysiological information about the brain networks involved in target behavior. Our results highlight the importance of individualized approaches for brain signal decoding, for which multielectrode recordings and spatial filtering can improve precision medicine approaches for clinical brain computer interfaces.
Collapse
Affiliation(s)
- Victoria Peterson
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| | - Timon Merk
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alan Bush
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Vadim Nikulin
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Andrea A Kühn
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Wolf-Julian Neumann
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
37
|
Lowet E, Kondabolu K, Zhou S, Mount RA, Wang Y, Ravasio CR, Han X. Deep brain stimulation creates informational lesion through membrane depolarization in mouse hippocampus. Nat Commun 2022; 13:7709. [PMID: 36513664 PMCID: PMC9748039 DOI: 10.1038/s41467-022-35314-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
Deep brain stimulation (DBS) is a promising neuromodulation therapy, but the neurophysiological mechanisms of DBS remain unclear. In awake mice, we performed high-speed membrane voltage fluorescence imaging of individual hippocampal CA1 neurons during DBS delivered at 40 Hz or 140 Hz, free of electrical interference. DBS powerfully depolarized somatic membrane potentials without suppressing spike rate, especially at 140 Hz. Further, DBS paced membrane voltage and spike timing at the stimulation frequency and reduced timed spiking output in response to hippocampal network theta-rhythmic (3-12 Hz) activity patterns. To determine whether DBS directly impacts cellular processing of inputs, we optogenetically evoked theta-rhythmic membrane depolarization at the soma. We found that DBS-evoked membrane depolarization was correlated with DBS-mediated suppression of neuronal responses to optogenetic inputs. These results demonstrate that DBS produces powerful membrane depolarization that interferes with the ability of individual neurons to respond to inputs, creating an informational lesion.
Collapse
Affiliation(s)
- Eric Lowet
- Boston University, Department of Biomedical Engineering, Boston, MA, 02215, USA.
| | - Krishnakanth Kondabolu
- grid.189504.10000 0004 1936 7558Boston University, Department of Biomedical Engineering, Boston, MA 02215 USA
| | - Samuel Zhou
- grid.189504.10000 0004 1936 7558Boston University, Department of Biomedical Engineering, Boston, MA 02215 USA
| | - Rebecca A. Mount
- grid.189504.10000 0004 1936 7558Boston University, Department of Biomedical Engineering, Boston, MA 02215 USA
| | - Yangyang Wang
- grid.189504.10000 0004 1936 7558Boston University, Department of Biomedical Engineering, Boston, MA 02215 USA
| | - Cara R. Ravasio
- grid.189504.10000 0004 1936 7558Boston University, Department of Biomedical Engineering, Boston, MA 02215 USA
| | - Xue Han
- Boston University, Department of Biomedical Engineering, Boston, MA, 02215, USA.
| |
Collapse
|
38
|
Takeuchi M, Tokutake K, Watanabe K, Ito N, Aoyama T, Saeki S, Kurimoto S, Hirata H, Hasegawa Y. A Wirelessly Powered 4-Channel Neurostimulator for Reconstructing Walking Trajectory. SENSORS (BASEL, SWITZERLAND) 2022; 22:7198. [PMID: 36236295 PMCID: PMC9572656 DOI: 10.3390/s22197198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
A wirelessly powered four-channel neurostimulator was developed for applying selective Functional Electrical Stimulation (FES) to four peripheral nerves to control the ankle and knee joints of a rat. The power of the neurostimulator was wirelessly supplied from a transmitter device, and the four nerves were connected to the receiver device, which controlled the ankle and knee joints in the rat. The receiver device had functions to detect the frequency of the transmitter signal from the transmitter coil. The stimulation site of the nerves was selected according to the frequency of the transmitter signal. The rat toe position was controlled by changing the angles of the ankle and knee joints. The joint angles were controlled by the stimulation current applied to each nerve independently. The stimulation currents were adjusted by the Proportional Integral Differential (PID) and feed-forward control method through a visual feedback control system, and the walking trajectory of a rat's hind leg was reconstructed. This study contributes to controlling the multiple joints of a leg and reconstructing functional motions such as walking using the robotic control technology.
Collapse
Affiliation(s)
- Masaru Takeuchi
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Katsuhiro Tokutake
- Department of Human Enhancement and Hand Surgery, Nagoya University, Nagoya 464-8601, Japan
| | - Keita Watanabe
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Naoyuki Ito
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Tadayoshi Aoyama
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Sota Saeki
- Department of Human Enhancement and Hand Surgery, Nagoya University, Nagoya 464-8601, Japan
| | - Shigeru Kurimoto
- Department of Human Enhancement and Hand Surgery, Nagoya University, Nagoya 464-8601, Japan
| | - Hitoshi Hirata
- Department of Human Enhancement and Hand Surgery, Nagoya University, Nagoya 464-8601, Japan
| | - Yasuhisa Hasegawa
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
39
|
Tan H. Towards adaptive deep brain stimulation for freezing of gait. Brain 2022; 145:2236-2238. [PMID: 35848860 PMCID: PMC9337803 DOI: 10.1093/brain/awac172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Huiling Tan
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
Legarda SB, Michas-Martin PA, McDermott D. Managing Intractable Symptoms of Parkinson's Disease: A Nonsurgical Approach Employing Infralow Frequency Neuromodulation. Front Hum Neurosci 2022; 16:894781. [PMID: 35880105 PMCID: PMC9308006 DOI: 10.3389/fnhum.2022.894781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
|
41
|
Cassar IR, Grill WM. The cortical evoked potential corresponds with deep brain stimulation efficacy in rats. J Neurophysiol 2022; 127:1253-1268. [PMID: 35389751 PMCID: PMC9054265 DOI: 10.1152/jn.00353.2021] [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: 08/05/2021] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 01/21/2023] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) antidromically activates the motor cortex (M1), and this cortical activation appears to play a role in the treatment of hypokinetic motor behaviors (Gradinaru V, Mogri M, Thompson KR, Henderson JM, Deisseroth K. Science 324: 354-359, 2009; Yu C, Cassar IR, Sambangi J, Grill WM. J Neurosci 40: 4323-4334, 2020). The synchronous antidromic activation takes the form of a short-latency cortical evoked potential (cEP) in electrocorticography (ECoG) recordings of M1. We assessed the utility of the cEP as a biomarker for STN DBS in unilateral 6-hydroxydopamine-lesioned female Sprague Dawley rats, with stimulating electrodes implanted in the STN and the ECoG recorded above M1. We quantified the correlations of the cEP magnitude and latency with changes in motor behavior from DBS and compared them to the correlation between motor behaviors and several commonly used spectral-based biomarkers. The cEP features correlated strongly with motor behaviors and were highly consistent across animals, whereas the spectral biomarkers correlated weakly with motor behaviors and were highly variable across animals. The cEP may thus be a useful biomarker for assessing the therapeutic efficacy of DBS parameters, as its features strongly correlate with motor behavior, it is consistent across time and subjects, it can be recorded under anesthesia, and it is simple to quantify with a large signal-to-noise ratio, enabling rapid, real-time evaluation. Additionally, our work provides further evidence that antidromic cortical activation mediates changes in motor behavior from STN DBS and that the dependence of DBS efficacy on stimulation frequency may be related to antidromic spike failure.NEW & NOTEWORTHY We characterize a new potential biomarker for deep brain stimulation (DBS), the cortical evoked potential (cEP), and demonstrate that it exhibits a robust correlation with motor behaviors as a function of stimulation frequency. The cEP may thus be a useful clinical biomarker for changes in motor behavior. This work also provides insight into the cortical mechanisms of DBS, suggesting that motor behaviors are strongly affected by the rate of antidromic spike failure during DBS.
Collapse
Affiliation(s)
- Isaac R Cassar
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina
- Department of Neurobiology, Duke University, Durham, North Carolina
- Department of Neurosurgery, Duke University, Durham, North Carolina
| |
Collapse
|
42
|
Silverio AA, Silverio LAA. Developments in Deep Brain Stimulators for Successful Aging Towards Smart Devices—An Overview. FRONTIERS IN AGING 2022; 3:848219. [PMID: 35821845 PMCID: PMC9261350 DOI: 10.3389/fragi.2022.848219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/15/2022] [Indexed: 12/02/2022]
Abstract
This work provides an overview of the present state-of-the-art in the development of deep brain Deep Brain Stimulation (DBS) and how such devices alleviate motor and cognitive disorders for a successful aging. This work reviews chronic diseases that are addressable via DBS, reporting also the treatment efficacies. The underlying mechanism for DBS is also reported. A discussion on hardware developments focusing on DBS control paradigms is included specifically the open- and closed-loop “smart” control implementations. Furthermore, developments towards a “smart” DBS, while considering the design challenges, current state of the art, and constraints, are also presented. This work also showcased different methods, using ambient energy scavenging, that offer alternative solutions to prolong the battery life of the DBS device. These are geared towards a low maintenance, semi-autonomous, and less disruptive device to be used by the elderly patient suffering from motor and cognitive disorders.
Collapse
Affiliation(s)
- Angelito A. Silverio
- Department of Electronics Engineering, University of Santo Tomas, Manila, Philippines
- Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
- *Correspondence: Angelito A. Silverio,
| | | |
Collapse
|
43
|
Tanaka M, Yanagisawa T, Fukuma R, Tani N, Oshino S, Mihara M, Hattori N, Kajiyama Y, Hashimoto R, Ikeda M, Mochizuki H, Kishima H. Magnetoencephalography detects phase-amplitude coupling in Parkinson's disease. Sci Rep 2022; 12:1835. [PMID: 35115607 PMCID: PMC8813926 DOI: 10.1038/s41598-022-05901-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022] Open
Abstract
To characterize Parkinson's disease, abnormal phase-amplitude coupling is assessed in the cortico-basal circuit using invasive recordings. It is unknown whether the same phenomenon might be found in regions other than the cortico-basal ganglia circuit. We hypothesized that using magnetoencephalography to assess phase-amplitude coupling in the whole brain can characterize Parkinson's disease. We recorded resting-state magnetoencephalographic signals in patients with Parkinson's disease and in healthy age- and sex-matched participants. We compared whole-brain signals from the two groups, evaluating the power spectra of 3 frequency bands (alpha, 8-12 Hz; beta, 13-25 Hz; gamma, 50-100 Hz) and the coupling between gamma amplitude and alpha or beta phases. Patients with Parkinson's disease showed significant beta-gamma phase-amplitude coupling that was widely distributed in the sensorimotor, occipital, and temporal cortices; healthy participants showed such coupling only in parts of the somatosensory and temporal cortices. Moreover, beta- and gamma-band power differed significantly between participants in the two groups (P < 0.05). Finally, beta-gamma phase-amplitude coupling in the sensorimotor cortices correlated significantly with motor symptoms of Parkinson's disease (P < 0.05); beta- and gamma-band power did not. We thus demonstrated that beta-gamma phase-amplitude coupling in the resting state characterizes Parkinson's disease.
Collapse
Affiliation(s)
- Masataka Tanaka
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takufumi Yanagisawa
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Institute for Advanced Co-Creation Studies, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Neuroinformatics, ATR Computational Neuroscience Laboratories, 2-2-2 Hikaridai, Seika-cho, Kyoto, 619 0288, Japan.
| | - Ryohei Fukuma
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Neuroinformatics, ATR Computational Neuroscience Laboratories, 2-2-2 Hikaridai, Seika-cho, Kyoto, 619 0288, Japan
| | - Naoki Tani
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoru Oshino
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahito Mihara
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriaki Hattori
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Rehabilitation, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Yuta Kajiyama
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1 Ogawahigashi, Kodaira, Tokyo, 187-8553, Japan
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
44
|
del Campo-Vera RM, Tang AM, Gogia AS, Chen KH, Sebastian R, Gilbert ZD, Nune G, Liu CY, Kellis S, Lee B. Neuromodulation in Beta-Band Power Between Movement Execution and Inhibition in the Human Hippocampus. Neuromodulation 2022; 25:232-244. [PMID: 35125142 PMCID: PMC8727636 DOI: 10.1111/ner.13486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/08/2021] [Accepted: 06/01/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The hippocampus is thought to be involved in movement, but its precise role in movement execution and inhibition has not been well studied. Previous work with direct neural recordings has found beta-band (13-30 Hz) modulation in both movement execution and inhibition throughout the motor system, but the role of beta-band modulation in the hippocampus during movement inhibition is not well understood. Here, we perform a Go/No-Go reaching task in ten patients with medically refractory epilepsy to study human hippocampal beta-power changes during movement. MATERIALS AND METHODS Ten epilepsy patients (5 female; ages 21-46) were implanted with intracranial depth electrodes for seizure monitoring and localization. Local field potentials were sampled at 2000 Hz during a Go/No-Go movement task. Comparison of beta-band power between Go and No-Go conditions was conducted using Wilcoxon signed-rank hypothesis testing for each patient. Sub-analyses were conducted to assess differences in the anterior vs posterior contacts, ipsilateral vs contralateral contacts, and male vs female beta-power values. RESULTS Eight out of ten patients showed significant beta-power decreases during the Go movement response (p < 0.05) compared to baseline. Eight out of ten patients also showed significant beta-power increases in the No-Go condition, occurring in the absence of movement. No significant differences were noted between ipsilateral vs contralateral contacts nor in anterior vs posterior hippocampal contacts. Female participants had a higher task success rate than males and had significantly greater beta-power increases in the No-Go condition (p < 0.001). CONCLUSION These findings indicate that increases in hippocampal beta power are associated with movement inhibition. To the best of our knowledge, this study is the first to report this phenomenon in the human hippocampus. The beta band may represent a state-change signal involved in motor processing. Future focus on the beta band in understanding human motor and impulse control will be vital.
Collapse
Affiliation(s)
- Roberto Martin del Campo-Vera
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Austin M. Tang
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Angad S. Gogia
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Kuang-Hsuan Chen
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Rinu Sebastian
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Zachary D. Gilbert
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - George Nune
- Department of Neurology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States,USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States
| | - Charles Y. Liu
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States,USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States,Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Spencer Kellis
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States,USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States,Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States,Tianqiao and Chrissy Chen Brain-Machine Interface Center, Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, United States
| | - Brian Lee
- Department of Neurological Surgery, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States,USC Neurorestoration Center, Keck School of Medicine of USC, Los Angeles, CA, United States,Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
45
|
Pozzi NG, Isaias IU. Adaptive deep brain stimulation: Retuning Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:273-284. [PMID: 35034741 DOI: 10.1016/b978-0-12-819410-2.00015-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A brain-machine interface represents a promising therapeutic avenue for the treatment of many neurologic conditions. Deep brain stimulation (DBS) is an invasive, neuro-modulatory tool that can improve different neurologic disorders by delivering electric stimulation to selected brain areas. DBS is particularly successful in advanced Parkinson's disease (PD), where it allows sustained improvement of motor symptoms. However, this approach is still poorly standardized, with variable clinical outcomes. To achieve an optimal therapeutic effect, novel adaptive DBS (aDBS) systems are being developed. These devices operate by adapting stimulation parameters in response to an input signal that can represent symptoms, motor activity, or other behavioral features. Emerging evidence suggests greater efficacy with fewer adverse effects during aDBS compared with conventional DBS. We address this topic by discussing the basics principles of aDBS, reviewing current evidence, and tackling the many challenges posed by aDBS for PD.
Collapse
Affiliation(s)
- Nicoló G Pozzi
- Department of Neurology, University Hospital Würzburg and Julius Maximilian University Würzburg, Würzburg, Germany
| | - Ioannis U Isaias
- Department of Neurology, University Hospital Würzburg and Julius Maximilian University Würzburg, Würzburg, Germany.
| |
Collapse
|
46
|
Cosentino G, Todisco M, Blandini F. Noninvasive neuromodulation in Parkinson's disease: Neuroplasticity implication and therapeutic perspectives. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:185-198. [PMID: 35034733 DOI: 10.1016/b978-0-12-819410-2.00010-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Noninvasive brain stimulation techniques can be used to study in vivo the changes of cortical activity and plasticity in subjects with Parkinson's disease (PD). Also, an increasing number of studies have suggested a potential therapeutic effect of these techniques. High-frequency repetitive transcranial magnetic stimulation (rTMS) and anodal transcranial direct current stimulation (tDCS) represent the most used stimulation paradigms to treat motor and nonmotor symptoms of PD. Both techniques can enhance cortical activity, compensating for its reduction related to subcortical dysfunction in PD. However, the use of suboptimal stimulation parameters can lead to therapeutic failure. Clinical studies are warranted to clarify in PD the additional effects of these stimulation techniques on pharmacologic and neurorehabilitation treatments.
Collapse
Affiliation(s)
- Giuseppe Cosentino
- Translational Neurophysiology Research Unit, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Massimiliano Todisco
- Translational Neurophysiology Research Unit, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Movement Disorders Research Center, IRCCS Mondino Foundation, Pavia, Italy.
| | - Fabio Blandini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Movement Disorders Research Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
47
|
Abdul Nabi Ali A, Alam M, Klein SC, Behmann N, Krauss JK, Doll T, Blume H, Schwabe K. Predictive accuracy of CNN for cortical oscillatory activity in an acute rat model of parkinsonism. Neural Netw 2021; 146:334-340. [PMID: 34923220 DOI: 10.1016/j.neunet.2021.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/08/2021] [Accepted: 11/24/2021] [Indexed: 10/19/2022]
Abstract
In neurological and neuropsychiatric disorders neuronal oscillatory activity between basal ganglia and cortical circuits are altered, which may be useful as biomarker for adaptive deep brain stimulation. We investigated whether changes in the spectral power of oscillatory activity in the motor cortex (MCtx) and the sensorimotor cortex (SMCtx) of rats after injection of the dopamine (DA) receptor antagonist haloperidol (HALO) would be similar to those observed in Parkinson disease. Thereafter, we tested whether a convolutional neural network (CNN) model would identify brain signal alterations in this acute model of parkinsonism. A sixteen channel surface micro-electrocorticogram (ECoG) recording array was placed under the dura above the MCtx and SMCtx areas of one hemisphere under general anaesthesia in rats. Seven days after surgery, micro ECoG was recorded in individual free moving rats in three conditions: (1) basal activity, (2) after injection of HALO (0.5 mg/kg), and (3) with additional injection of apomorphine (APO) (1 mg/kg). Furthermore, a CNN-based classification consisting of 23,530 parameters was applied on the raw data. HALO injection decreased oscillatory theta band activity (4-8 Hz) and enhanced beta (12-30 Hz) and gamma (30-100 Hz) in MCtx and SMCtx, which was compensated after APO injection (P ¡ 0.001). Evaluation of classification performance of the CNN model provided accuracy of 92%, sensitivity of 90% and specificity of 93% on one-dimensional signals. The CNN proposed model requires a minimum of sensory hardware and may be integrated into future research on therapeutic devices for Parkinson disease, such as adaptive closed loop stimulation, thus contributing to more efficient way of treatment.
Collapse
Affiliation(s)
- Ali Abdul Nabi Ali
- Institute of Microelectronic Systems, Architectures and Systems, Leibniz University Hannover, Hannover, D-30167, Lower Saxony, Germany
| | - Mesbah Alam
- Department of Neurosurgery, Hannover Medical School, Hannover, D-30625, Lower Saxony, Germany.
| | - Simon C Klein
- Institute of Microelectronic Systems, Architectures and Systems, Leibniz University Hannover, Hannover, D-30167, Lower Saxony, Germany
| | - Nicolai Behmann
- Institute of Microelectronic Systems, Architectures and Systems, Leibniz University Hannover, Hannover, D-30167, Lower Saxony, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, D-30625, Lower Saxony, Germany
| | - Theodor Doll
- Biomaterial Engineering, Hannover Medical School and Translational Medical Engineering Fraunhofer ITEM, Hannover, D-30625, Lower Saxony, Germany
| | - Holger Blume
- Institute of Microelectronic Systems, Architectures and Systems, Leibniz University Hannover, Hannover, D-30167, Lower Saxony, Germany
| | - Kerstin Schwabe
- Department of Neurosurgery, Hannover Medical School, Hannover, D-30625, Lower Saxony, Germany
| |
Collapse
|
48
|
Tai CH. Subthalamic burst firing: A pathophysiological target in Parkinson's disease. Neurosci Biobehav Rev 2021; 132:410-419. [PMID: 34856222 DOI: 10.1016/j.neubiorev.2021.11.044] [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: 09/01/2021] [Revised: 11/16/2021] [Accepted: 11/28/2021] [Indexed: 11/27/2022]
Abstract
Understanding the pathophysiological mechanism of Parkinson's disease (PD) in the subthalamic nucleus (STN) has become a critical issue since deep brain stimulation (DBS) in this region has been proven as an effective treatment for this disease. The STN possesses a special ability to switch from the spike to the burst firing mode in response to dopamine deficiency in parkinsonism, and this STN burst is considered an electrophysiological signature of the cortico-basal ganglia circuit in the brains of PD patients. This review focuses on the role of STN burst firing in the pathophysiology of PD and during DBS. Here, we review existing literature on how STN bursts originate and the specific factors affecting their formation; how STN burst firing causes motor symptoms in PD and how interventions can rescue these symptoms. Finally, the similarities and differences between the two electrophysiological hallmarks of PD, STN burst firing and beta-oscillation, are discussed. STN burst firing should be considered as a pathophysiological target in PD during treatment with DBS.
Collapse
Affiliation(s)
- Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, No. 7, Jhongshan South Road, 100225, Taipei, Taiwan.
| |
Collapse
|
49
|
da Silva Lapa JD, Godinho FLF, Teixeira MJ, Listik C, Iglesio RF, Duarte KP, Cury RG. Should the Globus Pallidus Targeting Be Refined in Dystonia? J Neurol Surg A Cent Eur Neurosurg 2021; 83:361-367. [PMID: 34808675 DOI: 10.1055/s-0041-1735856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND STUDY AIMS Deep brain stimulation (DBS) of the globus pallidus internus (GPi) is a highly effective therapy for primary generalized and focal dystonias, but therapeutic success is compromised by a nonresponder rate of up to 20%. Variability in electrode placement and in tissue stimulated inside the GPi may explain in part different outcomes among patients. Refinement of the target within the pallidal area could be helpful for surgery planning and clinical outcomes. The objective of this study was to discuss current and potential methodological (somatotopy, neuroimaging, and neurophysiology) aspects that might assist neurosurgical targeting of the GPi, aiming to treat generalized or focal dystonia. METHODS We selected published studies by searching electronic databases and scanning the reference lists for articles that examined the anatomical and electrophysiologic aspects of the GPi in patients with idiopathic/inherited dystonia who underwent functional neurosurgical procedures. RESULTS The sensorimotor sector of the GPi was the best target to treat dystonic symptoms, and was localized at its lateral posteroventral portion. The effective volume of tissue activated (VTA) to treat dystonia had a mean volume of 153 mm3 in the posterior GPi area. Initial tractography studies evaluated the close relation between the electrode localization and pallidothalamic tract to control dystonic symptoms.Regarding the somatotopy, the more ventral, lateral, and posterior areas of the GPi are associated with orofacial and cervical representation. In contrast, the more dorsal, medial, and anterior areas are associated with the lower limbs; between those areas, there is the representation of the upper limb. Excessive pallidal synchronization has a peak at the theta band of 3 to 8 Hz, which might be responsible for generating dystonic symptoms. CONCLUSIONS Somatotopy assessment of posteroventral GPi contributes to target-specific GPi sectors related to segmental body symptoms. Tractography delineates GPi output pathways that might guide electrode implants, and electrophysiology might assist in pointing out areas of excessive theta synchronization. Finally, the identification of oscillatory electrophysiologic features that correlate with symptoms might enable closed-loop approaches in the future.
Collapse
Affiliation(s)
- Jorge Dornellys da Silva Lapa
- Neurosurgery Unit, Fundação de Beneficiência Hospital de Cirurgia, Cirurgia, Aracaju, Sergipe, Brazil.,Division of Functional Neurosurgery, Department of Neurology, University of São Paulo, School of Medicine, Sao Paulo, São Paulo, Brazil
| | - Fábio Luiz Franceschi Godinho
- Division of Functional Neurosurgery, Department of Neurology, University of São Paulo, School of Medicine, Sao Paulo, São Paulo, Brazil
| | | | - Clarice Listik
- Movement Disorders Center, Department of Neurology, School of Medicine, University of Sao Paulo, Sao Paulo, São Paulo, Brazil
| | - Ricardo Ferrareto Iglesio
- Division of Functional Neurosurgery, Department of Neurology, University of São Paulo, School of Medicine, Sao Paulo, São Paulo, Brazil
| | - Kleber Paiva Duarte
- Division of Functional Neurosurgery, Department of Neurology, University of São Paulo, School of Medicine, Sao Paulo, São Paulo, Brazil
| | - Rubens Gisbert Cury
- Movement Disorders Center, Department of Neurology, School of Medicine, University of Sao Paulo, Sao Paulo, São Paulo, Brazil
| |
Collapse
|
50
|
di Biase L, Tinkhauser G, Martin Moraud E, Caminiti ML, Pecoraro PM, Di Lazzaro V. Adaptive, personalized closed-loop therapy for Parkinson's disease: biochemical, neurophysiological, and wearable sensing systems. Expert Rev Neurother 2021; 21:1371-1388. [PMID: 34736368 DOI: 10.1080/14737175.2021.2000392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Motor complication management is one of the main unmet needs in Parkinson's disease patients. AREAS COVERED Among the most promising emerging approaches for handling motor complications in Parkinson's disease, adaptive deep brain stimulation strategies operating in closed-loop have emerged as pivotal to deliver sustained, near-to-physiological inputs to dysfunctional basal ganglia-cortical circuits over time. Existing sensing systems that can provide feedback signals to close the loop include biochemical-, neurophysiological- or wearable-sensors. Biochemical sensing allows to directly monitor the pharmacokinetic and pharmacodynamic of antiparkinsonian drugs and metabolites. Neurophysiological sensing relies on neurotechnologies to sense cortical or subcortical brain activity and extract real-time correlates of symptom intensity or symptom control during DBS. A more direct representation of the symptom state, particularly the phenomenological differentiation and quantification of motor symptoms, can be realized via wearable sensor technology. EXPERT OPINION Biochemical, neurophysiologic, and wearable-based biomarkers are promising technological tools that either individually or in combination could guide adaptive therapy for Parkinson's disease motor symptoms in the future.
Collapse
Affiliation(s)
- Lazzaro di Biase
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico Di Roma, Rome, Italy.,Brain Innovations Lab, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Gerd Tinkhauser
- Department of Neurology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Eduardo Martin Moraud
- Department of Clinical Neurosciences, Lausanne University Hospital (Chuv) and University of Lausanne (Unil), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (.neurorestore), Lausanne University Hospital and Swiss Federal Institute of Technology (Epfl), Lausanne, Switzerland
| | - Maria Letizia Caminiti
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Pasquale Maria Pecoraro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico Di Roma, Rome, Italy
| |
Collapse
|