1
|
Boutom SM, Silva TP, Palecek SP, Shusta EV, Fernandes TG, Ashton RS. Central nervous system vascularization in human embryos and neural organoids. Cell Rep 2024; 43:115068. [PMID: 39693224 DOI: 10.1016/j.celrep.2024.115068] [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: 05/07/2024] [Revised: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
In recent years, neural organoids derived from human pluripotent stem cells (hPSCs) have offered a transformative pre-clinical platform for understanding central nervous system (CNS) development, disease, drug effects, and toxicology. CNS vasculature plays an important role in all these scenarios; however, most published studies describe CNS organoids that lack a functional vasculature or demonstrate rudimentary incorporation of endothelial cells or blood vessel networks. Here, we review the existing knowledge of vascularization during the development of different CNS regions, including the brain, spinal cord, and retina, and compare it to vascularized CNS organoid models. We highlight several areas of contrast where further bioengineering innovation is needed and discuss potential applications of vascularized neural organoids in modeling human CNS development, physiology, and disease.
Collapse
Affiliation(s)
- Sarah M Boutom
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Teresa P Silva
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tiago G Fernandes
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Randolph S Ashton
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Lin K, Stiles J, Tambo W, Ajmal E, Piao Q, Powell K, Li C. Bimodal functions of calcitonin gene-related peptide in the brain. Life Sci 2024; 359:123177. [PMID: 39486618 DOI: 10.1016/j.lfs.2024.123177] [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: 08/23/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
AIMS Calcitonin gene-related peptide (CGRP) is a pluripotent neuropeptide crucial for maintaining vascular homeostasis, yet its full therapeutic potential remains incompletely exploited. Within the brain, CGRP demonstrates a distinct bimodal effect, contributing to neuroprotection in ischemic conditions while inducing neuronal sensitization and inflammation in non-ischemic settings. Despite extensive research on CGRP, the absence of a definitive determinant for this observed dichotomy has limited its potential for therapeutic applications in the brain. This review examines the effects of CGRP in both physiological and pathological conditions, aiming to identify a unifying factor that could enhance its therapeutic applicability. MATERIALS AND METHODS This comprehensive literature review analyzes the molecular pathways associated with CGRP and the specific cellular responses observed in these contexts. Additionally, the review investigates the psychological implications of CGRP in relation to cerebral perfusion levels, aiming to elucidate its underlying factors. KEY FINDINGS Reviewing the literature reveals that, elevated levels of CGRP in non-ischemic conditions exert detrimental effects on brain function, while they confer protective effects in the context of ischemia. These encompass anti-oxidative, anti-inflammatory, anti-apoptotic, and angiogenic properties, along with behavioral normalization. Current findings indicate promising therapeutic avenues for CGRP beyond the acute phases of cerebral injury, extending to neurodegenerative and psychological disorders associated with cerebral hypoperfusion, as well as chronic recovery following acute cerebral injuries. SIGNIFICANCE Improved understanding of CGRP's bimodal properties, alongside advancements in CGRP delivery methodologies and brain ischemia detection technologies, paves the way for realizing its untapped potential and broad therapeutic benefits in diverse pathological conditions.
Collapse
Affiliation(s)
- Kanheng Lin
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Emory University, Atlanta, GA, USA
| | - Jacob Stiles
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; The College of William & Mary, Williamsburg, VA, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Erum Ajmal
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, USA
| | - Quanyu Piao
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
3
|
Peltonen S, Sonninen TM, Niskanen J, Koistinaho J, Ruponen M, Lehtonen Š. Mutated LRRK2 induces a reactive phenotype and alters migration in human iPSC-derived pericyte-like cells. Fluids Barriers CNS 2024; 21:92. [PMID: 39551752 PMCID: PMC11571670 DOI: 10.1186/s12987-024-00592-y] [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: 09/17/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Pericytes play a crucial role in controlling inflammation and vascular functions in the central nervous system, which are disrupted in Parkinson's disease (PD). Still, there is a lack of studies on the impact of pericytes on neurodegenerative diseases, and their involvement in the pathology of PD is unclear. Our objective was to investigate the molecular and functional differences between healthy pericytes and pericytes with the LRRK2 G2019S mutation, which is one of the most common mutations associated with PD. METHODS Our study employed pericyte-like cells obtained from induced pluripotent stem cells produced from PD patients with the LRRK2 G2019S mutation as well as from healthy individuals. We examined the gene expression profiles of the cells and analyzed how the alterations reflect on their functionality. RESULTS We have shown differences in the expression of genes related to inflammation and angiogenesis. Furthermore, we observe modified migration speed in PD pericyte-like cells as well as enhanced secretion of inflammatory mediators, such as soluble VCAM-1 and MCP-1, in these pericyte-like cells following exposure to proinflammatory stimuli. CONCLUSIONS In summary, our findings support the notion that pericytes play a role in the inflammatory and vascular changes observed in PD. Further investigation of pericytes could provide valuable insight into understanding the pathogenesis of PD.
Collapse
Affiliation(s)
- Sanni Peltonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tuuli-Maria Sonninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jonna Niskanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
- Neuroscience Center, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
4
|
Qiu J, Peng S, Qu R, Wu L, Xing L, Zhang L, Sun J. New evidence of vascular defects in neurodegenerative diseases revealed by single cell RNA sequencing. Clin Sci (Lond) 2024; 138:1377-1394. [PMID: 39469930 DOI: 10.1042/cs20241658] [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: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Neurodegenerative diseases (NDs) involve the progressive loss of neuronal structure or function in the brain and spinal cord. Despite their diverse etiologies, NDs manifest similar pathologies. Emerging research identifies vascular defects as a previously neglected hallmark of NDs. The development and popularization of single-cell RNA sequencing (scRNA-seq) technologies have significantly advanced our understanding of brain vascular cell types and their molecular characteristics, including gene expression changes at the single-cell level in NDs. These unprecedented insights deepen our understanding of the pathogenic mechanisms underlying NDs. However, the occurrence and role of vascular defects in disease progression remain largely unexplored. In this paper, we systematically summarize recent advances in the structure and organization of the central nervous system vasculature in mice, healthy individuals, and patients with NDs, focussing primarily on disease-specific alterations in vascular cell types or subtypes. Combining scRNA-seq with pathology evidence, we propose that vascular defects, characterized by disruptions in cell types and structural integrity, may serve as common early features of NDs. Finally, we discuss several pathways through which vascular defects in NDs lead to neuronal degeneration. A deeper understanding of the causes and contributions of vascular defects to NDs aids in elucidating the pathogenic mechanisms and developing meaningful therapeutic interventions.
Collapse
Affiliation(s)
- Jiaying Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
5
|
Chua J, Tan B, Wong D, Garhöfer G, Liew XW, Popa-Cherecheanu A, Loong Chin CW, Milea D, Li-Hsian Chen C, Schmetterer L. Optical coherence tomography angiography of the retina and choroid in systemic diseases. Prog Retin Eye Res 2024; 103:101292. [PMID: 39218142 DOI: 10.1016/j.preteyeres.2024.101292] [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: 05/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Optical coherence tomography angiography (OCTA) has transformed ocular vascular imaging, revealing microvascular changes linked to various systemic diseases. This review explores its applications in diabetes, hypertension, cardiovascular diseases, and neurodegenerative diseases. While OCTA provides a valuable window into the body's microvasculature, interpreting the findings can be complex. Additionally, challenges exist due to the relative non-specificity of its findings where changes observed in OCTA might not be unique to a specific disease, variations between OCTA machines, the lack of a standardized normative database for comparison, and potential image artifacts. Despite these limitations, OCTA holds immense potential for the future. The review highlights promising advancements like quantitative analysis of OCTA images, integration of artificial intelligence for faster and more accurate interpretation, and multi-modal imaging combining OCTA with other techniques for a more comprehensive characterization of the ocular vasculature. Furthermore, OCTA's potential future role in personalized medicine, enabling tailored treatment plans based on individual OCTA findings, community screening programs for early disease detection, and longitudinal studies tracking disease progression over time is also discussed. In conclusion, OCTA presents a significant opportunity to improve our understanding and management of systemic diseases. Addressing current limitations and pursuing these exciting future directions can solidify OCTA as an indispensable tool for diagnosis, monitoring disease progression, and potentially guiding treatment decisions across various systemic health conditions.
Collapse
Affiliation(s)
- Jacqueline Chua
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Bingyao Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore
| | - Damon Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Gerhard Garhöfer
- Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria
| | - Xin Wei Liew
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alina Popa-Cherecheanu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Emergency University Hospital, Department of Ophthalmology, Bucharest, Romania
| | - Calvin Woon Loong Chin
- Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Dan Milea
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Fondation Ophtalmologique Adolphe De Rothschild, Paris, France
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland; Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria; Fondation Ophtalmologique Adolphe De Rothschild, Paris, France; Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Strobel S, Sian-Hulsmann J, Tappe D, Jellinger K, Riederer P, Monoranu CM. Postencephalitic Parkinsonism: Unique Pathological and Clinical Features-Preliminary Data. Cells 2024; 13:1511. [PMID: 39329695 PMCID: PMC11430219 DOI: 10.3390/cells13181511] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Postencephalitic parkinsonism (PEP) is suggested to show a virus-induced pathology, which is different from classical idiopathic Parkinson's disease (PD) as there is no α-synuclein/Lewy body pathology. However, PEP shows a typical clinical representation of motor disturbances. In addition, compared to PD, there is no iron-induced pathology. The aim of this preliminary study was to compare PEP with PD regarding iron-induced pathology, using histochemistry methods on paraffin-embedded post-mortem brain tissue. In the PEP group, iron was not seen, except for one case with sparse perivascular depositions. Rather, PEP offers a pathology related to tau-protein/neurofibrillary tangles, with mild to moderate memory deficits only. It is assumed that this virus-induced pathology is due to immunological dysfunctions causing (neuro)inflammation-induced neuronal network disturbances as events that trigger clinical parkinsonism. The absence of iron deposits implies that PEP cannot be treated with iron chelators. The therapy with L-Dopa is also not an option, as L-Dopa only leads to an initial slight improvement in symptoms in isolated cases.
Collapse
Affiliation(s)
- Sabrina Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Jeswinder Sian-Hulsmann
- Department of Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi 00100, Kenya;
| | - Dennis Tappe
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Kurt Jellinger
- Institute of Clinical Neurobiology, A-1150 Vienna, Austria;
| | - Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, 97080 Wuerzburg, Germany;
- Department of Psychiatry, University of Southern Denmark, 5000 Odense, Denmark
| | - Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, 97080 Wuerzburg, Germany;
| |
Collapse
|
7
|
Zhang Y, Li Y, Lai C, Wang X, Luo F, Niu Y, Qin Y, Zhang Y, Zhang H. Interocular asymmetry of retinal change in Parkinson's disease. Parkinsonism Relat Disord 2024; 125:107037. [PMID: 38878445 DOI: 10.1016/j.parkreldis.2024.107037] [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: 01/09/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 07/29/2024]
Abstract
PURPOSE To investigate interocular asymmetry (IA) of retinal structure and vessel density in patients with Parkinson's disease (PD) and normal controls (NC). METHODS Seventy-eight subjects including 40 PD patients and 38 NC had completed optical coherence tomography angiography (OCTA) and neurological examinations for three rating scales (UPDRS-III, MMSE and MoCA). The IA was calculated by the absolute value of difference in right and left eyes. The IA of thickness in macular ganglion cell inner-plexiform layer (GCIPL), peripapillary retinal nerve fiber layer (pRNFL), and vessel density of superficial capillary plexus (SCP), deep capillary plexus (DCP), radial peripapillary capillary (RPC) were obtained from OCTA. RESULTS The motor-symptom-onset side of eyes showed lower vessel density in parafovea of SCP (51.09 ± 3.46 vs 49.81 ± 4.16, P = 0.03) and superior hemi of perifovea DCP (49.55 ± 5.81 vs 47.33 ± 5.71, = 0.04). The PD patients showed larger IA in thickness of superior half of pRNFL (5.27 [2.67, 10.87] vs 3.08 [1.62, 5.61], P = 0.02) and parafovea GCIPL (2.40[1.25, 6.35] vs 1.40[0.50, 2.45], P = 0.02). No significant interocular asymmetry was found in vessel density between PD and NC. A higher UPDRS-III scale was associated with larger IA in GCIPL (β = 0.093, P = 0.001) and smaller IA in DCP (β = -0.065, P = 0.037). CONCLUSION The motor-symptom-onset side of eyes showed more severe loss of macular vessel density than the other side of eyes. The PD patients showed asymmetrical structural change in GCIPL and pRNFL, which showed the potential as the diagnostic biomarker for PD.
Collapse
Affiliation(s)
- Yuqiao Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yan Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chunxin Lai
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital. Guangzhou, China; Shantou University Medical College, Shantou, China
| | - Xinhao Wang
- Department of Neurology, Maoming People's Hospital, Maoming, China
| | - Fulong Luo
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital. Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yongyi Niu
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital. Guangzhou, China
| | - Yongjie Qin
- Department of Ophthalmology, Guangdong Eye Institute, Guangdong Provincial People's Hospital. Guangzhou, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Hongyang Zhang
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Richardson A, Kundu A, Henao R, Lee T, Scott BL, Grewal DS, Fekrat S. Multimodal Retinal Imaging Classification for Parkinson's Disease Using a Convolutional Neural Network. Transl Vis Sci Technol 2024; 13:23. [PMID: 39136960 PMCID: PMC11323992 DOI: 10.1167/tvst.13.8.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 06/23/2024] [Indexed: 08/16/2024] Open
Abstract
Purpose Changes in retinal structure and microvasculature are connected to parallel changes in the brain. Two recent studies described machine learning algorithms trained on retinal images and quantitative data that identified Alzheimer's dementia and mild cognitive impairment with high accuracy. Prior studies also demonstrated retinal differences in individuals with PD. Herein, we developed a convolutional neural network (CNN) to classify multimodal retinal imaging from either a Parkinson's disease (PD) or control group. Methods We trained a CNN to receive retinal image inputs of optical coherence tomography (OCT) ganglion cell-inner plexiform layer (GC-IPL) thickness color maps, OCT angiography 6 × 6-mm en face macular images of the superficial capillary plexus, and ultra-widefield (UWF) fundus color and autofluorescence photographs to classify the retinal imaging as PD or control. The model consists of a shared pretrained VGG19 feature extractor and image-specific feature transformations which converge to a single output. Model results were assessed using receiver operating characteristic (ROC) curves and bootstrapped 95% confidence intervals for area under the ROC curve (AUC) values. Results In total, 371 eyes of 249 control subjects and 75 eyes of 52 PD subjects were used for training, validation, and testing. Our best CNN variant achieved an AUC of 0.918. UWF color photographs were the most effective imaging input, and GC-IPL thickness maps were the least contributory. Conclusions Using retinal images, our pilot CNN was able to identify individuals with PD and serves as a proof of concept to spur the collection of larger imaging datasets needed for clinical-grade algorithms. Translational Relevance Developing machine learning models for automated detection of Parkinson's disease from retinal imaging could lead to earlier and more widespread diagnoses.
Collapse
Affiliation(s)
- Alexander Richardson
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
- Department of Computer Science, Duke University, Durham, NC, USA
| | - Anita Kundu
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Ricardo Henao
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
- Department of Computer Science, Duke University, Durham, NC, USA
| | - Terry Lee
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Burton L. Scott
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Dilraj S. Grewal
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Sharon Fekrat
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
9
|
Lau K, Kotzur R, Richter F. Blood-brain barrier alterations and their impact on Parkinson's disease pathogenesis and therapy. Transl Neurodegener 2024; 13:37. [PMID: 39075566 PMCID: PMC11285262 DOI: 10.1186/s40035-024-00430-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
There is increasing evidence for blood-brain barrier (BBB) alterations in Parkinson's disease (PD), the second most common neurodegenerative disorder with rapidly rising prevalence. Altered tight junction and transporter protein levels, accumulation of α-synuclein and increase in inflammatory processes lead to extravasation of blood molecules and vessel degeneration. This could result in a self-perpetuating pathophysiology of inflammation and BBB alteration, which contribute to neurodegeneration. Toxin exposure or α-synuclein over-expression in animal models has been shown to initiate similar pathologies, providing a platform to study underlying mechanisms and therapeutic interventions. Here we provide a comprehensive review of the current knowledge on BBB alterations in PD patients and how rodent models that replicate some of these changes can be used to study disease mechanisms. Specific challenges in assessing the BBB in patients and in healthy controls are discussed. Finally, a potential role of BBB alterations in disease pathogenesis and possible implications for therapy are explored. The interference of BBB alterations with current and novel therapeutic strategies requires more attention. Brain region-specific BBB alterations could also open up novel opportunities to target specifically vulnerable neuronal subpopulations.
Collapse
Affiliation(s)
- Kristina Lau
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Rebecca Kotzur
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
10
|
Chen T, Dai Y, Hu C, Lin Z, Wang S, Yang J, Zeng L, Li S, Li W. Cellular and molecular mechanisms of the blood-brain barrier dysfunction in neurodegenerative diseases. Fluids Barriers CNS 2024; 21:60. [PMID: 39030617 PMCID: PMC11264766 DOI: 10.1186/s12987-024-00557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Maintaining the structural and functional integrity of the blood-brain barrier (BBB) is vital for neuronal equilibrium and optimal brain function. Disruptions to BBB performance are implicated in the pathology of neurodegenerative diseases. MAIN BODY Early indicators of multiple neurodegenerative disorders in humans and animal models include impaired BBB stability, regional cerebral blood flow shortfalls, and vascular inflammation associated with BBB dysfunction. Understanding the cellular and molecular mechanisms of BBB dysfunction in brain disorders is crucial for elucidating the sustenance of neural computations under pathological conditions and for developing treatments for these diseases. This paper initially explores the cellular and molecular definition of the BBB, along with the signaling pathways regulating BBB stability, cerebral blood flow, and vascular inflammation. Subsequently, we review current insights into BBB dynamics in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The paper concludes by proposing a unified mechanism whereby BBB dysfunction contributes to neurodegenerative disorders, highlights potential BBB-focused therapeutic strategies and targets, and outlines lessons learned and future research directions. CONCLUSIONS BBB breakdown significantly impacts the development and progression of neurodegenerative diseases, and unraveling the cellular and molecular mechanisms underlying BBB dysfunction is vital to elucidate how neural computations are sustained under pathological conditions and to devise therapeutic approaches.
Collapse
Affiliation(s)
- Tongli Chen
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yan Dai
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Chenghao Hu
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Zihao Lin
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shengzhe Wang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jing Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Shanshan Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Weiyun Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
11
|
Samson JS, Ramesh A, Parvathi VD. Development of Midbrain Dopaminergic Neurons and the Advantage of Using hiPSCs as a Model System to Study Parkinson's Disease. Neuroscience 2024; 546:1-19. [PMID: 38522661 DOI: 10.1016/j.neuroscience.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Midbrain dopaminergic (mDA) neurons are significantly impaired in patients inflicted with Parkinson's disease (PD), subsequently affecting a variety of motor functions. There are four pathways through which dopamine elicits its function, namely, nigrostriatal, mesolimbic, mesocortical and tuberoinfundibular dopamine pathways. SHH and Wnt signalling pathways in association with favourable expression of a variety of genes, promotes the development and differentiation of mDA neurons in the brain. However, there is a knowledge gap regarding the complex signalling pathways involved in development of mDA neurons. hiPSC models have been acclaimed to be effective in generating complex disease phenotypes. These models mimic the microenvironment found in vivo thus ensuring maximum reliability. Further, a variety of therapeutic compounds can be screened using hiPSCs since they can be used to generate neurons that could carry an array of mutations associated with both familial and sporadic PD. Thus, culturing hiPSCs to study gene expression and dysregulation of cellular processes associated with PD can be useful in developing targeted therapies that will be a step towards halting disease progression.
Collapse
Affiliation(s)
- Jennifer Sally Samson
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Anuradha Ramesh
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai 600116, India.
| |
Collapse
|
12
|
Kanoh T, Mizoguchi T, Tonoki A, Itoh M. Modeling of age-related neurological disease: utility of zebrafish. Front Aging Neurosci 2024; 16:1399098. [PMID: 38765773 PMCID: PMC11099255 DOI: 10.3389/fnagi.2024.1399098] [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: 03/11/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
Many age-related neurological diseases still lack effective treatments, making their understanding a critical and urgent issue in the globally aging society. To overcome this challenge, an animal model that accurately mimics these diseases is essential. To date, many mouse models have been developed to induce age-related neurological diseases through genetic manipulation or drug administration. These models help in understanding disease mechanisms and finding potential therapeutic targets. However, some age-related neurological diseases cannot be fully replicated in human pathology due to the different aspects between humans and mice. Although zebrafish has recently come into focus as a promising model for studying aging, there are few genetic zebrafish models of the age-related neurological disease. This review compares the aging phenotypes of humans, mice, and zebrafish, and provides an overview of age-related neurological diseases that can be mimicked in mouse models and those that cannot. We presented the possibility that reproducing human cerebral small vessel diseases during aging might be difficult in mice, and zebrafish has potential to be another animal model of such diseases due to their similarity of aging phenotype to humans.
Collapse
Affiliation(s)
- Tohgo Kanoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Takamasa Mizoguchi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ayako Tonoki
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| |
Collapse
|
13
|
Shimizu F. [Blood-brain barrier breakdown and autoimmune cerebellar ataxia]. Rinsho Shinkeigaku 2024; 64:148-156. [PMID: 38403685 DOI: 10.5692/clinicalneurol.cn-001932] [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] [Indexed: 02/27/2024]
Abstract
Autoimmune cerebellar ataxia is a disease entity that affects the cerebellum and is induced by autoimmune mechanisms. The disease is classified into several etiologies, including gluten ataxia, anti-glutamate decarboxylase (GAD) ataxia, paraneoplastic cerebellar degeneration, primary autoimmune cerebellar ataxia and postinfectious cerebellar ataxia. The autoimmune response in the periphery cross-reacts with similar antigens in the cerebellum due to molecular mimicry. Breakdown of the blood‒brain barrier (BBB) could potentially explain the vulnerability of the cerebellum during the development of autoimmune cerebellar ataxia, as it gives rise to the entry of pathogenic autoantibodies or lymphocytes into the cerebellum. In this review, the maintenance of the BBB under normal conditions and the molecular basis of BBB disruption under pathological conditions are highlighted. Next, the pathomechanism of BBB breakdown in each subtype of autoimmune cerebellar ataxia is discussed. We recently identified glucose-regulated protein (GRP) 78 antibodies in paraneoplastic cerebellar degeneration and Lambert-Eaton myasthenic syndrome, and GRP78 antibodies induced by cross-reactivity with tumors can disrupt the BBB and penetrate anti-P/Q type voltage-gated calcium channel (VGCC) antibodies into the cerebellum, thus leading to cerebellar ataxia in this disease.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine
| |
Collapse
|
14
|
Wu C, Wu H, Zhou C, Guan X, Guo T, Wu J, Chen J, Wen J, Qin J, Tan S, Duanmu X, Yuan W, Zheng Q, Zhang B, Xu X, Zhang M. Neurovascular coupling alteration in drug-naïve Parkinson's disease: The underlying molecular mechanisms and levodopa's restoration effects. Neurobiol Dis 2024; 191:106406. [PMID: 38199273 DOI: 10.1016/j.nbd.2024.106406] [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: 11/12/2023] [Revised: 12/25/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) patients exhibit an imbalance between neuronal activity and perfusion, referred to as abnormal neurovascular coupling (NVC). Nevertheless, the underlying molecular mechanism and how levodopa, the standard treatment in PD, regulates NVC is largely unknown. MATERIAL AND METHODS A total of 52 drug-naïve PD patients and 49 normal controls (NCs) were enrolled. NVC was characterized in vivo by relating cerebral blood flow (CBF) and amplitude of low-frequency fluctuations (ALFF). Motor assessments and MRI scanning were conducted on drug-naïve patients before and after levodopa therapy (OFF/ON state). Regional NVC differences between patients and NCs were identified, followed by an assessment of the associated receptors/transporters. The influence of levodopa on NVC, CBF, and ALFF within these abnormal regions was analyzed. RESULTS Compared to NCs, OFF-state patients showed NVC dysfunction in significantly lower NVC in left precentral, postcentral, superior parietal cortex, and precuneus, along with higher NVC in left anterior cingulate cortex, right olfactory cortex, thalamus, caudate, and putamen (P-value <0.0006). The distribution of NVC differences correlated with the density of dopaminergic, serotonin, MU-opioid, and cholinergic receptors/transporters. Additionally, levodopa ameliorated abnormal NVC in most of these regions, where there were primarily ALFF changes with limited CBF modifications. CONCLUSION Patients exhibited NVC dysfunction primarily in the striato-thalamo-cortical circuit and motor control regions, which could be driven by dopaminergic and nondopaminergic systems, and levodopa therapy mainly restored abnormal NVC by modulating neuronal activity.
Collapse
Affiliation(s)
- Chenqing Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoting Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhou
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingwen Chen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Wen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmei Qin
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sijia Tan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojie Duanmu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijin Yuan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianshi Zheng
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
15
|
Holtman IR, Glass CK, Nott A. Interpretation of Neurodegenerative GWAS Risk Alleles in Microglia and their Interplay with Other Cell Types. ADVANCES IN NEUROBIOLOGY 2024; 37:531-544. [PMID: 39207711 DOI: 10.1007/978-3-031-55529-9_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia have been implicated in numerous neurodegenerative and neuroinflammatory disorders; however, the causal contribution of this immune cell type is frequently debated. Genetic studies offer a unique vantage point in that they infer causality over a secondary consequence. Genome-wide association studies (GWASs) have identified hundreds of loci in the genome that are associated with susceptibility to neurodegenerative disorders. GWAS studies implicate microglia in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and to a lesser degree suggest a role for microglia in vascular dementia (VaD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS), and other neurodegenerative and neuropsychiatric disorders. The contribution and function of GWAS risk loci on disease progression is an ongoing field of study, in which large genomic datasets, and an extensive framework of computational tools, have proven to be crucial. Several GWAS risk loci are shared between disorders, pointing towards common pleiotropic mechanisms. In this chapter, we introduce key concepts in GWAS and post-GWAS interpretation of neurodegenerative disorders, with a focus on GWAS risk genes implicated in microglia, their interplay with other cell types and shared convergence of GWAS risk loci on microglia.
Collapse
Affiliation(s)
- Inge R Holtman
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| |
Collapse
|
16
|
Wang J, Li H, Jia J, Shao X, Li Y, Zhou Y, Wang H, Jin L. Progressive Cerebrovascular Reactivity Reduction Occurs in Parkinson's Disease: A Longitudinal Study. Mov Disord 2024; 39:94-104. [PMID: 38013597 DOI: 10.1002/mds.29671] [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: 09/07/2023] [Revised: 10/15/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The change of microvascular function over the course of Parkinson's disease (PD) remains unclear. OBJECTIVE We aimed to ascertain regional cerebrovascular reactivity (CVR) changes in the patients with PD at baseline (V0) and during a 2-year follow-up period (V1). We further investigated whether alterations in CVR were linked to cognitive decline and brain functional connectivity (FC). METHODS We recruited 90 PD patients and 51 matched healthy controls (HCs). PD patients underwent clinical evaluations, neuropsychological assessments, and magnetic resonance (MR) scanning at V0 and V1, whereas HCs completed neuropsychological assessments and MR at baseline. The analysis included evaluating CVR and FC maps derived from resting-state functional magnetic resonance imaging and investigating CVR measurement reproducibility. RESULTS Compared with HCs, CVR reduction in left inferior occipital gyrus and right superior temporal cortex at V0 persisted at V1, with larger clusters. Longitudinal reduction in CVR of the left posterior cingulate cortex correlated with decline in Trail Making Test B performance within PD patients. Reproducibility validation further confirmed these findings. In addition, the results also showed that there was a tendency for FC to be weakened from posterior to anterior with the progression of the disease. CONCLUSIONS Microvascular dysfunction might be involved in disease progression, subsequently weaken brain FC, and partly contribute to executive function deficits in early PD. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jian Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University (Xiamen Branch), China
| | - Hongwei Li
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Jia Jia
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Neurology, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Xiali Shao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanfang Li
- Department of Neurology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Ying Zhou
- Department of Neurology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - He Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Lirong Jin
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Jeong JY, Lee HJ, Kim N, Li Y, Rah JC, Oh WJ. Impaired neuronal activity as a potential factor contributing to the underdeveloped cerebrovasculature in a young Parkinson's disease mouse model. Sci Rep 2023; 13:22613. [PMID: 38114623 PMCID: PMC10730707 DOI: 10.1038/s41598-023-49900-w] [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: 05/22/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Misfolding of α-synuclein (α-Syn) in the brain causes cellular dysfunction, leading to cell death in a group of neurons, and consequently causes the progression of Parkinson's disease (PD). Although many studies have demonstrated the pathological connections between vascular dysfunction and neurodegenerative diseases, it remains unclear how neuronal accumulation of α-Syn affects the structural and functional aspects of the cerebrovasculature to accelerate early disease progression. Here, we demonstrated the effect of aberrant α-Syn expression on the brain vasculature using a PD mouse model expressing a familial mutant form of human α-Syn selectively in neuronal cells. We showed that young PD mice have an underdeveloped cerebrovasculature without significant α-Syn accumulation in the vasculature. During the early phase of PD, toxic α-Syn was selectively increased in neuronal cells, while endothelial cell proliferation was decreased in the absence of vascular cell death or neuroinflammation. Instead, we observed altered neuronal activation and minor changes in the activity-dependent gene expression in brain endothelial cells (ECs) in young PD mice. These findings demonstrated that neuronal expression of mutant α-Syn in the early stage of PD induces abnormal neuronal activity and contributes to vascular patterning defects, which could be associated with a reduced angiogenic potential of ECs.
Collapse
Affiliation(s)
- Jin-Young Jeong
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, South Korea
| | - Hyun Jung Lee
- Sensory and Motor System Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Namsuk Kim
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Yan Li
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Jong-Cheol Rah
- Sensory and Motor System Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Won-Jong Oh
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea.
| |
Collapse
|
18
|
Castillo L, Berrozpe-Villabona C, Miserachs-García S, Haulani H, Gómez-Gutiérrez C, González-Martínez A, Morilla-Grasa A, Arias L, Caminal JM, Casaroli-Marano R. Choriocapillaris and choroidal thickness in all Leber hereditary optic neuropathy stages using swept source technology. Acta Ophthalmol 2023. [PMID: 37983892 DOI: 10.1111/aos.15811] [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: 04/25/2023] [Revised: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023]
Abstract
PURPOSE The role of the choroid in Leber hereditary optic neuropathy (LHON) remains unclear. The literature is scarce, with conflicting results and lacks axial length measurements. Therefore, we aimed to analyse the choriocapillaris (CC) vessel density (VD) and choroidal thickness (ChT) in all stages of LHON using swept source (SS) technology and considering the possible influence of axial length on choroidal parameters. METHODS This was a prospective cross-sectional observational study. A total of 119 eyes of 60 patients with molecularly confirmed LHON across all stages and 120 eyes of 60 control participants were included. We obtained the CC VD using optical coherence tomography angiography maps centred on the fovea. ChT was measured from the Bruch's membrane to the choroid-sclera interface in the macular and peripapillary regions. RESULTS The CC VD was not significantly affected in any sector or average, except for a slight change in the superior region of chronic eyes (52.08 ± 1.62% vs. 53.50 ± 2.29%, p = 0.002). ChT demonstrated a trend towards decreased values in asymptomatic eyes and increased values in the symptomatic stages that failed to reach statistical significance in sectors corresponding to the papillomacular bundle except for the macular nasal inner sector of chronic eyes (281.10 ± 67.12 μm vs. 252.08 ± 70.55 μm, p = 0.045). No significant correlations were observed between visual acuity and CC VD or ChT. CONCLUSION The CC VD remained stable across the LHON stages. Choroidal vasculature does not appear to play a role in LHON pathophysiology. Further research is needed on ChT as a potential biomarker of LHON.
Collapse
Affiliation(s)
| | - Clara Berrozpe-Villabona
- Department of Ophthalmology, Clínica Universidad de Navarra, Madrid, Spain
- Department of Ophthalmology, Clínica Universidad de Navarra, Pamplona, Spain
- Thematic Network of Cooperative Health Research in Eye Diseases (Oftared), Health Institute Carlos III, Madrid, Spain
| | - Sergio Miserachs-García
- Institut Clínic d'Oftalmologia (ICOF) - Seu Plató, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | | | | | | | - Luis Arias
- Department of Ophthalmology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - José M Caminal
- Department of Ophthalmology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ricardo Casaroli-Marano
- Department of Surgery, School of Medicine and Health Sciences, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Salehi MA, Rezagholi F, Mohammadi S, Zakavi SS, Jahanshahi A, Gouravani M, Yazdanpanah G, Seddon I, Jabbehdari S, Singh RP. Optical coherence tomography angiography measurements in Parkinson's disease: A systematic review and meta-analysis. Eye (Lond) 2023; 37:3145-3156. [PMID: 36941403 PMCID: PMC10564940 DOI: 10.1038/s41433-023-02483-2] [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: 08/23/2022] [Revised: 01/06/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
Optical coherence tomography angiography (OCT-A) is an ocular imaging technology that has emerged as a non-invasive tool to evaluate retinal microvascular changes in neurodegenerative diseases including Parkinson's disease (PD) and Alzheimer's disease. While several studies have reported on the presence of pathologic retinal microvascular alterations in PD, the utility of OCT-A as a biomarker for PD evaluation is still unclear. A systematic review and meta-analysis were performed to explore the current evidence for the role of OCT-A in PD published up until June 2022. PubMed, Scopus, and Web of Science databases were used to systematically identify relevant papers and a meta-analysis was conducted using Stata16 software according to the level of heterogeneity applying a random- or fixed-effect model. Thirteen studies of 925 eyes in the PD group and 1501 eyes in the control group assessing OCT-A findings in PD patients were included. The meta-analyses revealed that the foveal region of PD patients had a significantly lower vessel density in the superficial capillary plexus (SCP) compared to healthy controls but that there were no significant differences in the foveal avascular zone, the SCP in whole, parafoveal, and perifoveal regions, and deep capillary plexus. OCT-A metrics may act as a potential biomarker for a more accurate and early PD diagnosis. Still, the OCT-A algorithms and interchangeability between OCT-A devices require further standardization to draw clinical conclusions regarding their utility.
Collapse
Affiliation(s)
| | - Fateme Rezagholi
- School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Soheil Mohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Sina Zakavi
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Jahanshahi
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahdi Gouravani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Yazdanpanah
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Ian Seddon
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Sayena Jabbehdari
- Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Rishi P Singh
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
20
|
Joshi D, Prasad S, Saini J, Ingalhalikar M. Role of Arterial Spin Labeling (ASL) Images in Parkinson's Disease (PD): A Systematic Review. Acad Radiol 2023; 30:1695-1708. [PMID: 36435728 DOI: 10.1016/j.acra.2022.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022]
Abstract
RATIONALE AND OBJECTIVES Parkinson's disease is a chronic progressive neurodegenerative disorder with standard structural MRIs often showing no gross abnormalities. Quantitative perfusion MRI modality Arterial Spin Labeling (ASL) is helpful in identifying PD specific perfusion patterns. Absolute Cerebral blood flow (CBF) measurement using ASL provides insights into regional perfusion abnormalities. We reviewed the role of ASL to identify specific brain regions responsible for motor, non-motor symptoms and neurovascular changes observed in PD. Challenges in assessing the blood perfusion level are discussed with future development for improving the evaluation of ASL perfusion maps. MATERIALS AND METHODS We included CBF quantification studies using ASL for PD diagnosis. A systematic search was performed in Pubmed, Scopus and Web of Science. The perfusion parameters CBF and arterial arrival time (AAT) measured using ASL were considered for brain region assessment. Clinical aspects of PD have been analyzed using ASL perfusion maps. RESULTS The systematic search identified 153 unique records. Thirty articles were selected after verification of inclusion and exclusion criteria. Voxel and region-based analyses in white and gray matter tissues have been performed to identify PD-specific perfusion patterns by reported articles. Predominant brain regions such as basal ganglia sub-regions, frontoparietal network, precuneus, occipital lobe, sensory motor area regions, visual network, which are associated with motor and non-motor symptoms in PD, were identified with CBF hypoperfusion, indicating neuronal loss and cerebrovascular dysfunction. CONCLUSION CBF and AAT values derived from ASL can potentially be used as biomarkers to discriminate PD from similar brain-related disorders.
Collapse
Affiliation(s)
- Dhanashri Joshi
- Symbiosis Center of Medical Image Analysis, Symbiosis International (Deemed) University, Pune,MH, India
| | - Shweta Prasad
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, KA, India; Department of Clinical Neurosciences, National Institute of Mental Health and Neurosciences, Bengaluru,, KA, India
| | - Jitender Saini
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health and Neurosciences, Bengaluru, KA, India
| | - Madhura Ingalhalikar
- Symbiosis Center of Medical Image Analysis, Symbiosis International (Deemed) University, Pune,MH, India.
| |
Collapse
|
21
|
Pamphlett R, Bishop DP. The toxic metal hypothesis for neurological disorders. Front Neurol 2023; 14:1173779. [PMID: 37426441 PMCID: PMC10328356 DOI: 10.3389/fneur.2023.1173779] [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: 02/25/2023] [Accepted: 05/30/2023] [Indexed: 07/11/2023] Open
Abstract
Multiple sclerosis and the major sporadic neurogenerative disorders, amyotrophic lateral sclerosis, Parkinson disease, and Alzheimer disease are considered to have both genetic and environmental components. Advances have been made in finding genetic predispositions to these disorders, but it has been difficult to pin down environmental agents that trigger them. Environmental toxic metals have been implicated in neurological disorders, since human exposure to toxic metals is common from anthropogenic and natural sources, and toxic metals have damaging properties that are suspected to underlie many of these disorders. Questions remain, however, as to how toxic metals enter the nervous system, if one or combinations of metals are sufficient to precipitate disease, and how toxic metal exposure results in different patterns of neuronal and white matter loss. The hypothesis presented here is that damage to selective locus ceruleus neurons from toxic metals causes dysfunction of the blood-brain barrier. This allows circulating toxicants to enter astrocytes, from where they are transferred to, and damage, oligodendrocytes, and neurons. The type of neurological disorder that arises depends on (i) which locus ceruleus neurons are damaged, (ii) genetic variants that give rise to susceptibility to toxic metal uptake, cytotoxicity, or clearance, (iii) the age, frequency, and duration of toxicant exposure, and (iv) the uptake of various mixtures of toxic metals. Evidence supporting this hypothesis is presented, concentrating on studies that have examined the distribution of toxic metals in the human nervous system. Clinicopathological features shared between neurological disorders are listed that can be linked to toxic metals. Details are provided on how the hypothesis applies to multiple sclerosis and the major neurodegenerative disorders. Further avenues to explore the toxic metal hypothesis for neurological disorders are suggested. In conclusion, environmental toxic metals may play a part in several common neurological disorders. While further evidence to support this hypothesis is needed, to protect the nervous system it would be prudent to take steps to reduce environmental toxic metal pollution from industrial, mining, and manufacturing sources, and from the burning of fossil fuels.
Collapse
Affiliation(s)
- Roger Pamphlett
- Department of Pathology, Brain and Mind Centre, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - David P. Bishop
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Mármol I, Abizanda-Campo S, Ayuso JM, Ochoa I, Oliván S. Towards Novel Biomimetic In Vitro Models of the Blood-Brain Barrier for Drug Permeability Evaluation. Bioengineering (Basel) 2023; 10:bioengineering10050572. [PMID: 37237642 DOI: 10.3390/bioengineering10050572] [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: 04/18/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Current available animal and in vitro cell-based models for studying brain-related pathologies and drug evaluation face several limitations since they are unable to reproduce the unique architecture and physiology of the human blood-brain barrier. Because of that, promising preclinical drug candidates often fail in clinical trials due to their inability to penetrate the blood-brain barrier (BBB). Therefore, novel models that allow us to successfully predict drug permeability through the BBB would accelerate the implementation of much-needed therapies for glioblastoma, Alzheimer's disease, and further disorders. In line with this, organ-on-chip models of the BBB are an interesting alternative to traditional models. These microfluidic models provide the necessary support to recreate the architecture of the BBB and mimic the fluidic conditions of the cerebral microvasculature. Herein, the most recent advances in organ-on-chip models for the BBB are reviewed, focusing on their potential to provide robust and reliable data regarding drug candidate ability to reach the brain parenchyma. We point out recent achievements and challenges to overcome in order to advance in more biomimetic in vitro experimental models based on OOO technology. The minimum requirements that should be met to be considered biomimetic (cellular types, fluid flow, and tissular architecture), and consequently, a solid alternative to in vitro traditional models or animals.
Collapse
Affiliation(s)
- Inés Mármol
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Sara Abizanda-Campo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Department of Dermatology, Department of Biomedical Engineering, and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jose M Ayuso
- Department of Dermatology, Department of Biomedical Engineering, and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
23
|
Central retinal microvasculature damage is associated with orthostatic hypotension in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:36. [PMID: 36894544 PMCID: PMC9998652 DOI: 10.1038/s41531-023-00480-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Orthostatic hypotension (OH) is a common non-motor symptom in Parkinson's disease (PD). OH can cause cerebral and retinal hypoperfusion and is associated with microvascular damage in PD. Optical coherence tomography angiography (OCTA) is a non-invasive technology that can be used to visualize the retinal microvasculature and detect microvascular damage in PD. In the present study, 51 PD patients (OH+, n = 20, 37 eyes; OH-, n = 32, 61 eyes) and 51 healthy controls (100 eyes) were evaluated. The Unified Parkinson's Disease Rating Scale III, Hoehn and Yahr scale, Montreal Cognitive Assessment, levodopa equivalent daily dose, and vascular risk factors, including hypertension, diabetes, and dyslipidemia, were investigated. PD patients underwent a head-up tilt (HUT) test. The PD patients had a lower superficial retinal capillary plexus (SRCP) density in the central region than control patients. The PDOH+ group had lower vessel density in the SRCP of the central region compared with the control group and lower vessel density in the DRCP of the central region than the PDOH- and control groups. The changes in systolic and diastolic blood pressure during the HUT test in PD patients showed a negative correlation with the vessel density in the DRCP central region. The presence of OH was a critical factor associated with central microvasculature damage in PD. These findings indicate that OCTA can be a useful and non-invasive tool for detecting microvasculature damage in PD patients.
Collapse
|
24
|
Elabi OF, Karampatsi D, Vercalsteren E, Lietzau G, Nyström T, Klein T, Darsalia V, Patrone C, Paul G. DPP-4 Inhibitor and Sulfonylurea Differentially Reverse Type 2 Diabetes-Induced Blood-Brain Barrier Leakage and Normalize Capillary Pericyte Coverage. Diabetes 2023; 72:405-414. [PMID: 36448982 PMCID: PMC9935496 DOI: 10.2337/db22-0674] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022]
Abstract
Microvascular pathology in the brain is one of the suggested mechanisms underlying the increased incidence and progression of neurodegenerative diseases in people with type 2 diabetes (T2D). Although accumulating data suggest a neuroprotective effect of antidiabetics, the underlying mechanisms are unclear. Here, we investigated whether two clinically used antidiabetics, the dipeptidyl peptidase-4 inhibitor linagliptin and the sulfonylurea glimepiride, which restore T2D-induced brain vascular pathology. Microvascular pathology was examined in the striatum of mice fed for 12 months with either normal chow diet or a high-fat diet (HFD) to induce T2D. A subgroup of HFD-fed mice was treated with either linagliptin or glimepiride for 3 months before sacrifice. We demonstrate that T2D caused leakage of the blood-brain barrier (BBB), induced angiogenesis, and reduced pericyte coverage of microvessels. However, linagliptin and glimepiride recovered the BBB integrity and restored the pericyte coverage differentially. Linagliptin normalized T2D-induced angiogenesis and restored pericyte coverage. In contrast, glimepiride enhanced T2D-induced angiogenesis and increased pericyte density, resulting in proper vascular coverage. Interestingly, glimepiride reduced microglial activation, increased microglial-vascular interaction, and increased collagen IV density. This study provides evidence that both DPP-4 inhibition and sulfonylurea reverse T2D-induced BBB leakage, which may contribute to antidiabetic neurorestorative effects.
Collapse
Affiliation(s)
- Osama F. Elabi
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Dimitra Karampatsi
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ellen Vercalsteren
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Grazyna Lietzau
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Vladimer Darsalia
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cesare Patrone
- NeuroCardioMetabol Group, Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
- Corresponding author: Gesine Paul,
| |
Collapse
|
25
|
Stroke in Parkinson's disease: a review of epidemiological studies and potential pathophysiological mechanisms. Acta Neurol Belg 2023:10.1007/s13760-023-02202-4. [PMID: 36710306 DOI: 10.1007/s13760-023-02202-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is the fastest growing neurological disorder and one of the leading neurological causes of disability worldwide following stroke. An overall aging global population, as well as general changes in lifestyle associated with mass industrialization in the last century, may be linked to both increased incidence rates of PD and an increase in cumulative cardiovascular risk. Recent epidemiological studies show an increased risk of stroke, post-stroke complications, and subclinical ischemic insults in PD. PD patients have a host of characteristics that might contribute to increasing the risk of developing ischemic stroke including motor impairment, dysautonomia, and sleep disorders. This increases the urgency to study the interplay between PD and other neurological disorders, and their combined effect on mortality, morbidity, and quality of life. In this review, we provide a comprehensive overview of the studied etiological factors and pathological processes involved in PD, specifically with regard to their relationship to stroke. We hope that this review offers an insight into the relationship between PD and ischemic stroke and motivates further studies in this regard.
Collapse
|
26
|
Schurhoff N, Toborek M. Circadian rhythms in the blood-brain barrier: impact on neurological disorders and stress responses. Mol Brain 2023; 16:5. [PMID: 36635730 PMCID: PMC9835375 DOI: 10.1186/s13041-023-00997-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Circadian disruption has become more prevalent in society due to the increase in shift work, sleep disruption, blue light exposure, and travel via different time zones. The circadian rhythm is a timed transcription-translation feedback loop with positive regulators, BMAL1 and CLOCK, that interact with negative regulators, CRY and PER, to regulate both the central and peripheral clocks. This review highlights the functions of the circadian rhythm, specifically in the blood-brain barrier (BBB), during both healthy and pathological states. The BBB is a highly selective dynamic interface composed of CNS endothelial cells, astrocytes, pericytes, neurons, and microglia that form the neurovascular unit (NVU). Circadian rhythms modulate BBB integrity through regulating oscillations of tight junction proteins, assisting in functions of the NVU, and modulating transporter functions. Circadian disruptions within the BBB have been observed in stress responses and several neurological disorders, including brain metastasis, epilepsy, Alzheimer's disease, and Parkinson's disease. Further understanding of these interactions may facilitate the development of improved treatment options and preventative measures.
Collapse
Affiliation(s)
- Nicolette Schurhoff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065, Katowice, Poland.
| |
Collapse
|
27
|
Zhang D, Fu Q, Xue C, Xiao C, Sun Y, Liu W, Hu X. Characterization of Hemodynamic Alteration in Parkinson's Disease and Effect on Resting-State Connectivity. Neuroscience 2023:S0306-4522(23)00006-4. [PMID: 36642395 DOI: 10.1016/j.neuroscience.2023.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023]
Abstract
Functional magnetic resonance imaging (fMRI) is a convolution of latent neural activity and the hemodynamic response function (HRF). According to prior studies, the neurodegenerative process in idiopathic Parkinson's Disease (PD) interacts significantly with neuromuscular abnormalities. Although these underlying neuromuscular changes might influence the temporal characteristics of HRF and fMRI signals, relatively few studies have explored this possibility. We hypothesized that such alterations would engender changes in estimated functional connectivity (FC) in fMRI space compared to latent neural space. To test these theories, we calculated voxel-level HRFs by deconvolving resting-state fMRI data from PD patients (n = 61) and healthy controls (HC) (n = 47). Significant group differences in HRF (P < 0.05, Gaussian random field-corrected) were observed in several regions previously associated with PD. Subsequently, we focused on putamen-seed-based FC differences between the PD and HC groups using fMRI and latent neural signals. The results suggested that neglecting HRF variability may cultivate false-positive and false-negative FC group differences. Furthermore, HRF was related to dopamine receptor type 2 (DRD2) gene expression (P < 0.001, t = -7.06, false discover rate-corrected). Taken together, these findings reveal HRF variation and its possible underlying molecular mechanism in PD, and suggest that deconvolution could reduce the impact of HRF variation on FC group differences.
Collapse
Affiliation(s)
- Da Zhang
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianyi Fu
- International Laboratory for Children's Medical Imaging Research, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, Jiangsu, China
| | - Chen Xue
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chaoyong Xiao
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Sun
- International Laboratory for Children's Medical Imaging Research, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, Jiangsu, China; Research Centre for University of Birmingham and Southeast University, Southeast University, Nanjing, Jiangsu, China
| | - Weiguo Liu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiao Hu
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Institute of Brain Functional Imaging, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Blasiak J, Sobczuk P, Pawlowska E, Kaarniranta K. Interplay between aging and other factors of the pathogenesis of age-related macular degeneration. Ageing Res Rev 2022; 81:101735. [PMID: 36113764 DOI: 10.1016/j.arr.2022.101735] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 01/31/2023]
Abstract
Age-related macular degeneration (AMD) is a complex eye disease with the retina as the target tissue and aging as per definition the most serious risk factor. However, the retina contains over 60 kinds of cells that form different structures, including the neuroretina and retinal pigment epithelium (RPE) which can age at different rates. Other established or putative AMD risk factors can differentially affect the neuroretina and RPE and can differently interplay with aging of these structures. The occurrence of β-amyloid plaques and increased levels of cholesterol in AMD retinas suggest that AMD may be a syndrome of accelerated brain aging. Therefore, the question about the real meaning of age in AMD is justified. In this review we present and update information on how aging may interplay with some aspects of AMD pathogenesis, such as oxidative stress, amyloid beta formation, circadian rhythm, metabolic aging and cellular senescence. Also, we show how this interplay can be specific for photoreceptors, microglia cells and RPE cells as well as in Bruch's membrane and the choroid. Therefore, the process of aging may differentially affect different retinal structures. As an accurate quantification of biological aging is important for risk stratification and early intervention for age-related diseases, the determination how photoreceptors, microglial and RPE cells age in AMD may be helpful for a precise diagnosis and treatment of this largely untreatable disease.
Collapse
Affiliation(s)
- Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| | - Piotr Sobczuk
- Emergency Medicine and Disaster Medicine Department, Medical University of Lodz, Pomorska 251, 92-209 Lodz, Poland; Department of Orthopaedics and Traumatology, Polish Mothers' Memorial Hospital - Research Institute, Rzgowska 281, 93-338 Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, Pomorska 251, 92-216 Lodz, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, KYS, P.O. Box 100, FI-70029 Finland
| |
Collapse
|
29
|
Cesaroni V, Blandini F, Cerri S. Dyskinesia and Parkinson's disease: animal model, drug targets, and agents in preclinical testing. Expert Opin Ther Targets 2022; 26:837-851. [PMID: 36469635 DOI: 10.1080/14728222.2022.2153036] [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] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. PD patients exhibit a classic spectrum of motor symptoms, arising when dopamine neurons in the substantia nigra pars compacta are reduced by 60%. The dopamine precursor L-DOPA represents the most effective therapy for improving PD motor dysfunctions, thus far available. Unfortunately, long-term treatment with L-DOPA is associated with the development of severe side effects, resulting in abnormal involuntary movements termed levodopa-induced dyskinesia (LID). Amantadine is the only drug currently approved for the treatment of LID indicating that LID management is still an unmet need in PD and encouraging the search for novel anti-dyskinetic drugs or the assessment of combined therapies with different molecular targets. AREAS COVERED This review provides an overview of the main preclinical models used to study LID and of the latest preclinical evidence on experimental and clinically available pharmacological approaches targeting non-dopaminergic systems. EXPERT OPINION LIDs are supported by complex molecular and neurobiological mechanisms that are still being studied today. This complexity suggests the need of developing personalized pharmacological approach to obtain an effective amelioration of LID condition and improve the quality of life of PD patients.
Collapse
Affiliation(s)
- Valentina Cesaroni
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation 27100, Pavia, Italy
| | - Fabio Blandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico 20122, Milan, Italy
| | - Silvia Cerri
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation 27100, Pavia, Italy
| |
Collapse
|