1
|
Medina R, Derias AM, Lakdawala M, Speakman S, Lucke-Wold B. Overview of emerging therapies for demyelinating diseases. World J Clin Cases 2024; 12:6361-6373. [PMID: 39464332 PMCID: PMC11438674 DOI: 10.12998/wjcc.v12.i30.6361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
This paper provides an overview of autoimmune disorders of the central nervous system, specifically those caused by demyelination. We explore new research regarding potential therapeutic interventions, particularly those aimed at inducing remyelination. Remyelination is a detailed process, involving many cell types-oligodendrocyte precursor cells (OPCs), astrocytes, and microglia-and both the innate and adaptive immune systems. Our discussion of this process includes the differentiation potential of neural stem cells, the function of adult OPCs, and the impact of molecular mediators on myelin repair. Emerging therapies are also explored, with mechanisms of action including the induction of OPC differentiation, the transplantation of mesenchymal stem cells, and the use of molecular mediators. Further, we discuss current medical advancements in relation to many myelin-related disorders, including multiple sclerosis, optic neuritis, neuromyelitis optica spectrum disorder, myelin oligodendrocyte glycoprotein antibody-associated disease, transverse myelitis, and acute disseminated encephalomyelitis. Beyond these emerging systemic therapies, we also introduce the dimethyl fumarate/silk fibroin nerve conduit and its potential role in the treatment of peripheral nerve injuries. Despite these aforementioned scientific advancements, this paper maintains the need for ongoing research to deepen our understanding of demyelinating diseases and advance therapeutic strategies that enhance affected patients' quality of life.
Collapse
Affiliation(s)
- Robert Medina
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Ann-Marie Derias
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Maria Lakdawala
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Skye Speakman
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
2
|
Peters A, Banine F, Yasuhara K, Hoffman A, Basappa, Metri PK, Gunning L, Huffman A, VanCampen J, Shock CC, Back SA, Sherman LS. Distinct chemical structures inhibit the CEMIP hyaluronidase and promote oligodendrocyte progenitor cell maturation. J Biol Chem 2024:107916. [PMID: 39454959 DOI: 10.1016/j.jbc.2024.107916] [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: 02/28/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Growing evidence supports pathogenic roles for chronically elevated hyaluronidase activity in numerous conditions. Elevated expression of one such hyaluronidase, the Cell Migration Inducing and hyaluronan binding Protein (CEMIP), has been implicated in the pathogenesis and progression of several cancers as well as demyelinating diseases in the central nervous system (CNS). Developing effective and selective CEMIP inhibitors could therefore have efficacy in treating a variety of conditions where CEMIP is chronically elevated. Using two distinct screens for novel hyaluronidase inhibitors, we identified two synthetic thiocarbamates and one plant-derived flavonoid, sulfuretin, that effectively blocked CEMIP activity in live cells, including a tumorigenic cell line and primary cultures of oligodendrocyte progenitor cells (OPCs). None of these agents influenced cell proliferation, but they had differential dose-dependent and cell type specific effects on cell survival. Furthermore, we found that each of these agents could promote oligodendrocyte maturation by OPCs in the presence of high molecular weight (>2 Mda) hyaluronan, the accumulation of which is linked to the inhibition of OPC maturation and remyelination failure in demyelinating diseases. These findings indicate that CEMIP can be inhibited through distinct chemical interactions, and that CEMIP inhibitors have potential efficacy for treating demyelinating diseases or other conditions where CEMIP is elevated.
Collapse
Affiliation(s)
- Alec Peters
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Fatima Banine
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Kanon Yasuhara
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Angela Hoffman
- Department of Chemistry, University of Portland, Portland OR 97203, USA
| | - Basappa
- Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, India
| | - Prashant K Metri
- Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, India
| | - Lily Gunning
- Department of Chemistry, University of Portland, Portland OR 97203, USA
| | - Ava Huffman
- Department of Chemistry, University of Portland, Portland OR 97203, USA
| | - Jake VanCampen
- Department of Chemistry, University of Portland, Portland OR 97203, USA
| | - Clinton C Shock
- College of Agricultural Sciences, Oregon State University, Corvallis, OR 97331
| | - Stephen A Back
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
3
|
Schrod S, Lück N, Lohmayer R, Solbrig S, Völkl D, Wipfler T, Shutta KH, Ben Guebila M, Schäfer A, Beißbarth T, Zacharias HU, Oefner PJ, Quackenbush J, Altenbuchinger M. Spatial Cellular Networks from omics data with SpaCeNet. Genome Res 2024; 34:1371-1383. [PMID: 39231609 PMCID: PMC11529864 DOI: 10.1101/gr.279125.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Advances in omics technologies have allowed spatially resolved molecular profiling of single cells, providing a window not only into the diversity and distribution of cell types within a tissue, but also into the effects of interactions between cells in shaping the transcriptional landscape. Cells send chemical and mechanical signals which are received by other cells, where they can subsequently initiate context-specific gene regulatory responses. These interactions and their responses shape the individual molecular phenotype of a cell in a given microenvironment. RNAs or proteins measured in individual cells, together with the cells' spatial distribution, provide invaluable information about these mechanisms and the regulation of genes beyond processes occurring independently in each individual cell. "SpaCeNet" is a method designed to elucidate both the intracellular molecular networks (how molecular variables affect each other within the cell) and the intercellular molecular networks (how cells affect molecular variables in their neighbors). This is achieved by estimating conditional independence (CI) relations between captured variables within individual cells and by disentangling these from CI relations between variables of different cells.
Collapse
Affiliation(s)
- Stefan Schrod
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Niklas Lück
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Robert Lohmayer
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Stefan Solbrig
- Institute of Theoretical Physics, University of Regensburg, 93053 Regensburg, Germany
| | - Dennis Völkl
- Institute of Theoretical Physics, University of Regensburg, 93053 Regensburg, Germany
| | - Tina Wipfler
- Institute of Theoretical Physics, University of Regensburg, 93053 Regensburg, Germany
| | - Katherine H Shutta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Andreas Schäfer
- Institute of Theoretical Physics, University of Regensburg, 93053 Regensburg, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany
- Campus Institute Data Science (CIDAS), University of Göttingen, 37077 Göttingen, Germany
| | - Helena U Zacharias
- Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Hannover Medical School, 30625 Hannover, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Michael Altenbuchinger
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37077 Göttingen, Germany;
| |
Collapse
|
4
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
5
|
Tang W, Wang Q, Sun M, Liu C, Huang Y, Zhou M, Zhang X, Meng Z, Zhang J. The gut microbiota-oligodendrocyte axis: A promising pathway for modulating oligodendrocyte homeostasis and demyelination-associated disorders. Life Sci 2024; 354:122952. [PMID: 39127317 DOI: 10.1016/j.lfs.2024.122952] [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: 04/23/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
The bidirectional regulation between the gut microbiota and brain, known as gut-brain axis, has received significant attention. The myelin sheath, produced by oligodendrocytes or Schwann cells, is essential for efficient nervous signal transmission and the maintenance of brain function. Growing evidence shows that both oligodendrogenesis and myelination are modulated by gut microbiota and its metabolites, and when dysbiosis occurs, changes in the microbiota composition and/or associated metabolites may impact developmental myelination and the occurrence of neurodevelopmental disabilities. Although the link between the microbiota and demyelinating disease such as multiple sclerosis has been extensively studied, our knowledge about the role of the microbiota in other myelin-related disorders, such as neurodegenerative diseases, is limited. Mechanistically, the microbiota-oligodendrocyte axis is primarily mediated by factors such as inflammation, the vagus nerve, endocrine hormones, and microbiota metabolites as evidenced by metagenomics, metabolomics, vagotomy, and morphological and molecular approaches. Treatments targeting this axis include probiotics, prebiotics, microbial metabolites, herbal bioactive compounds, and specific dietary management. In addition to the commonly used approaches, viral vector-mediated tracing and gene manipulation, integrated multiomics and multicenter clinical trials will greatly promote the mechanistic and interventional studies and ultimately, the development of new preventive and therapeutic strategies against gut-oligodendrocyte axis-mediated brain impairments. Interestingly, recent findings showed that microbiota dysbiosis can be induced by hippocampal myelin damage and is reversible by myelin-targeted drugs, which provides new insights into understanding how hippocampus-based functional impairment (such as in neurodegenerative Alzheimer's disease) regulates the peripheral homeostasis of microbiota and associated systemic disorders.
Collapse
Affiliation(s)
- Wen Tang
- Department of Gastroenterology, Chongqing Western Hospital, Chongqing 400052, China
| | - Qi Wang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Mingguang Sun
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China; Department of Neurology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing University of Chinese Medicine, Beijing 100853, China
| | - Chang''e Liu
- Department of Nutrition, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Yonghua Huang
- Department of Neurology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Maohu Zhou
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Xuan Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Zhaoyou Meng
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
6
|
Dadkhah M, Afshari S, Samizadegan T, Shirmard LR, Barin S. Pegylated chitosan nanoparticles of fluoxetine enhance cognitive performance and hippocampal brain derived neurotrophic factor levels in a rat model of local demyelination. Exp Gerontol 2024; 195:112533. [PMID: 39134215 DOI: 10.1016/j.exger.2024.112533] [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: 05/15/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024]
Abstract
Cognitive impairment is a common feature in neurodegenerative diseases such as multiple sclerosis (MS). This study aims to explore the potential of enhancing the beneficial effects of fluoxetine (FLX), a neuroprotective agent known for its ability to increase neural plasticity by utilizing nanoparticles. The study specifically focuses on the synthesis and evaluation of PEGylated chitosan nanoparticles of FLX and its effect on demyelination and the subsequent cognitive impairment (CI) in the hippocampus of rats induced by local injection of lysophosphatidylcholine (LPC). Chitosan/polyethylene glycol nanoparticles were synthesized, and their properties were analyzed. Demyelination was induced in rats via hippocampal injections of lysolecithin. Behavioral assessments included open field maze, elevated plus maze, and novel object recognition memory (NORM) tests. Hippocampal levels of insulin-like growth factor (IGF-1) and brain-derived neurotrophic factor (BDNF) were measured using enzyme-linked immunoassay (ELISA). The extent of remyelination was quantified using Luxol fast blue staining. Nanoparticle size measured 240.2 nm with 53 % encapsulation efficacy. Drug release exhibited a slow pattern, with 76 % released within 4 h. Nanoparticle-treated rats displayed reduced anxiety-like behavior, improved memory, increased BDNF levels, and a reduced extent of demyelination, with no change in IGF- levels. In addition, FLX -loaded chitosan nanoparticles had better effect on cognitive improvement, BDNF levels in the hippocampus that FLX. Altering pharmacokinetics and possibly pharmacodynamics. These findings highlight the potential of innovative drug delivery systems, encouraging further research in this direction.
Collapse
Affiliation(s)
- Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Salva Afshari
- Student Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Tara Samizadegan
- Student Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Leila Rezaie Shirmard
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Sajjad Barin
- Department of Pathology, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
7
|
Almuklass AM, Alassaf AM, Alanazi RF, Alnafisah TR, Alrehaily TA, Al Malik Y. Nerve conduction, latency, and its association with hand function in young men. PLoS One 2024; 19:e0310813. [PMID: 39348363 PMCID: PMC11441660 DOI: 10.1371/journal.pone.0310813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/07/2024] [Indexed: 10/02/2024] Open
Abstract
INTRODUCTION The median and ulnar nerves have been suggested to play a significant role in hand function; however, there are insufficient data to determine the strength of this association. This study aimed to investigate the correlation between hand function as measured with the Grooved pegboard test (GPT) and conduction velocity and latency of the median and ulnar nerves. METHODS We collected convenience samples in the College of Medicine, KSAU-HS. We used GPT to characterize hand function and performed measured nerve conduction velocity (NCV) and latency of the ulnar and median nerves of both hands. We used the Edinburgh handedness inventory (EHI) to determine hand dominance. RESULTS We recruited 28 healthy medical students aged 20-29 years (mean: 21.46 ± 1.62 years). Most were right-handed (n = 25, 89.3%), with a mean EHI score of 302 ± 210. The mean GPT time was significantly faster in the dominant (65.5 ± 6.4 s) than in the non-dominant (75.0 ± 9.6 s) hand. The NCV for the ulnar nerve of the dominant hand was significantly correlated with GPT (r = -0.52, p = 0.005) while median nerve was not correlated (0.24, p = 0.21). Regression analysis and collinearity test showed that the ulnar NCV explained 20% of the variance in GPT of the dominant hand (R2 = 0.203, p = 0.016). CONCLUSION The ulnar nerve conduction velocity, explained 20% of the variance in GPT times of the young men. Performance on this biomarker of neurological health seems to be more influenced by other factors in healthy young individuals.
Collapse
Affiliation(s)
- Awad M Almuklass
- Basic Medical Science Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Abdulmajeed Mansour Alassaf
- Basic Medical Science Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Rakan F Alanazi
- Basic Medical Science Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Turki Rashed Alnafisah
- Basic Medical Science Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Thamir Ali Alrehaily
- Basic Medical Science Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Yaser Al Malik
- Basic Medical Science Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Division of Neurology, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Wei X, Liu S, Chen K, Wang M, Wang Y, Zou D, Xiao Y. Knockdown of BMP7 induced oligodendrocyte apoptosis, demyelination and motor function loss. Mol Cell Neurosci 2024; 131:103973. [PMID: 39332617 DOI: 10.1016/j.mcn.2024.103973] [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: 07/16/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Demyelinating diseases, including multiple sclerosis (MS) and spinal cord injury (SCI), lead to significant neurological deficits primarily due to the loss of oligodendrocytes (OLs). Bone Morphogenetic Protein 7 (BMP7) is expressed abundantly in the central nervous system and previous studies showed its protective effect in reducing OL loss. In this study, we aim to explore BMP7's potential as a biomarker and therapeutic target for demyelinating diseases by investigating its expression and effects on OLs and myelin sheath integrity. METHOD We analyzed multiple Gene Expression Omnibus datasets for BMP7 expression profiles in demyelinating conditions such as MS and SCI. Experimentally, we employed a BMP7 knockdown model in rat spinal cords using adeno-associated virus8 vectors to specifically reduce BMP7 expression. Western blotting, immunofluorescence, and Nissl staining were used to assess the effect on OL and other types of cells. The structure of myelin sheath and locomotor function were evaluated using transmission electron microscopy and BBB scores, and statistical analysis included ROC curves and ANOVA to evaluate BMP7's diagnostic and therapeutic potential. RESULTS BMP7 expression consistently decreased across various demyelinating models, and BMP7 knockdown led to increased OL apoptosis through the Smad1/5/9 pathway, with no apparent effect on other cell types. This reduction in OLs was associated with myelin degeneration, axonal damage, and impaired motor function. CONCLUSION The study confirms BMP7's significant involvement in the pathophysiology of demyelinating diseases and supports its potential as a therapeutic target or biomarker. Future research should focus on therapeutic strategies to enhance BMP7 function and further investigate the mechanisms by which BMP7 supports myelin integrity.
Collapse
Affiliation(s)
- Xiaojin Wei
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuxin Liu
- Department of Pain Management and Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kai Chen
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan, China.
| | - Meng Wang
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan, China
| | - Yaping Wang
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan, China.
| | - Dingquan Zou
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan, China.
| | - Yanying Xiao
- Department of Pain Management and Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
9
|
Yang L, Pan W, Cai Q, An M, Wang C, Pan X. The research trend on neurobrucellosis over the past 30 years (1993-2023): a bibliometric and visualization analysis. Front Neurol 2024; 15:1349530. [PMID: 39381075 PMCID: PMC11460295 DOI: 10.3389/fneur.2024.1349530] [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: 12/13/2023] [Accepted: 09/02/2024] [Indexed: 10/10/2024] Open
Abstract
Background Brucellosis is a zoonotic disease caused by Brucella infection, which is common in pastoral areas. Neurological involvement in brucellosis is relatively rare. But since 1993, continuous studies have been reporting neurological complications of brucellosis, collectively referred to as neurobrucellosis. A bibliometric analysis of existing literature outlines current research progress and gaps and provides guidance for the clinical treatment of neurobrucellosis, promoting patient health in the process of guiding clinical practice, and improving their quality of life. Methods CiteSpace and VOSviewer are software tools to visualize research trends and networks. By selecting specific areas of interest and configuring the right parameters, the tools can visualize past research data. The study retrieved the literature from the Web of Science Core Collection Database and downloaded it in plain text file format. Citespace6.1.6, VOSviewer v1.6.20, and Microsoft Excel 16.59 were used for analyzing the following terms: countries, institutions, authors' cooperation, journals, keywords, and co-citation. Results There are eight key results. (1) The publication volume shows a general upward trend. (2) Turkey is the country with the highest publication volume and contributing institutions. (3) Giambartolomei GH, Gul HC, and Namiduru M are the authors with the highest number of publications. (4) Neurology is the journal that published the highest number of related articles (n = 12). (5) "Diagnosis," "meningitis," and "features" are the top three frequently occurring keywords. (6) Keyword clusters show "antibiotic therapy" and "cerebrospinal fluid" have future study value. (7) The burst analysis of the keywords also indicates that "cerebrospinal fluid" may become a prominent keyword in future research. (8) The co-citation analysis concludes three categories of the cited articles, which are diagnosis, therapy, and complications, indicating the past research direction. Conclusion This study highlights the complexity of neurobrucellosis, presenting the need for advanced diagnostic techniques and multifaceted treatment approaches. While antibiotics remain the cornerstone of therapy, the use of corticosteroids to mitigate inflammatory responses shows promise, albeit with concerns about potential sequelae and relapse. Future research should focus on refining therapeutic strategies that address both the direct effects of infection and the broader immunological impacts to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Lanting Yang
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
- Faculty of Arts, The University of Melbourne, Melbourne, VIC, Australia
| | - Wei Pan
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Qiong Cai
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Mingyang An
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Chunjie Wang
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Xilong Pan
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
10
|
Kushwaha S, Saji J, Verma R, Singh V, Ansari JA, Mishra SK, Roy O, Patnaik S, Ghosh D. Microglial Neuroinflammation-Independent Reversal of Demyelination of Corpus Callosum by Arsenic in a Cuprizone-Induced Demyelinating Mouse Model. Mol Neurobiol 2024; 61:6822-6841. [PMID: 38353925 DOI: 10.1007/s12035-024-03978-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: 08/31/2023] [Accepted: 01/19/2024] [Indexed: 08/22/2024]
Abstract
Demyelination is the loss of myelin in CNS, resulting in damaged myelin sheath. Oxidative stress and neuroinflammation play a key role in inducing demyelinating diseases like MS; hence, controlling oxidative stress and neuroinflammation is important. Cuprizone (CPZ), a copper chelator, generates oxidative stress and neuroinflammation, thereby inducing demyelination. Therefore, the CPZ-induced demyelinating mouse model (CPZ model) is widely used in research. The present study was intended to unravel a mechanism of inhibition of demyelination by arsenic in a CPZ model, which is otherwise known for its toxicity. We investigated an alternative mechanism of inhibition of demyelination by arsenic through the reversal of SOD1 activity employing in silico analysis, analytical chemistry techniques, and in vitro and in vivo experiments. In vivo experiments showed protection of body weight, survivability, and myelination of the corpus callosum in CPZ and arsenic-co-exposed animals, where neuroinflammation was apparently not involved. In vitro experiments revealed that arsenic-mediated reversal of impaired SOD1 activity leads to reduced cellular ROS levels and better viability of primary oligodendrocytes. Reversal of SOD1 activity was also observed in the corpus callosum tissue isolated from experimental animals. In silico and analytical chemistry studies revealed that similar to copper, arsenic can potentially bind to CPZ and thereby make the copper freely available for SOD1 activity. Suitable neurobehavior tests further validated the protective effect of arsenic. Taken together, the present study revealed that arsenic protects oligodendrocytes and demyelination of corpus callosum by reversing CPZ-induced impaired SOD1 activity.
Collapse
Affiliation(s)
- Shaivya Kushwaha
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Joel Saji
- Water Analysis Laboratory, Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, 226001, India
| | - Rahul Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Water Analysis Laboratory, Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, 226001, India
| | - Vikas Singh
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jamal Ahmad Ansari
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Genome Instability and Chromatin Remodeling Section, NIH-National Institute of Aging, Baltimore, USA
| | - Shubhendra Kumar Mishra
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Department of Pediatrics, Division of Neonatology, McGill University Health Centre-Research Institute (RI-MUHC), Montreal, QC, Canada
| | - Opalina Roy
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
| | - Satyakam Patnaik
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Water Analysis Laboratory, Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, 226001, India.
| | - Debabrata Ghosh
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
11
|
Khodanovich M, Svetlik M, Kamaeva D, Usova A, Kudabaeva M, Anan’ina T, Vasserlauf I, Pashkevich V, Moshkina M, Obukhovskaya V, Kataeva N, Levina A, Tumentceva Y, Vasilieva S, Schastnyy E, Naumova A. Demyelination in Patients with POST-COVID Depression. J Clin Med 2024; 13:4692. [PMID: 39200834 PMCID: PMC11355865 DOI: 10.3390/jcm13164692] [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: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Depression is one of the most severe sequelae of COVID-19, with major depressive disorder often characterized by disruption in white matter (WM) connectivity stemming from changes in brain myelination. This study aimed to quantitatively assess brain myelination in clinically diagnosed post-COVID depression (PCD) using the recently proposed MRI method, macromolecular proton fraction (MPF) mapping. Methods: The study involved 63 recovered COVID-19 patients (52 mild, 11 moderate, and 2 severe) at 13.5 ± 10.0 months post-recovery, with matched controls without prior COVID-19 history (n = 19). A post-COVID depression group (PCD, n = 25) was identified based on psychiatric diagnosis, while a comparison group (noPCD, n = 38) included participants with neurological COVID-19 complications, excluding clinical depression. Results: Fast MPF mapping revealed extensive demyelination in PCD patients, particularly in juxtacortical WM (predominantly occipital lobe and medial surface), WM tracts (inferior fronto-occipital fasciculus (IFOF), posterior thalamic radiation, external capsule, sagittal stratum, tapetum), and grey matter (GM) structures (hippocampus, putamen, globus pallidus, and amygdala). The noPCD group also displayed notable demyelination, but with less magnitude and propagation. Multiple regression analysis highlighted IFOF demyelination as the primary predictor of Hamilton scores, PCD presence, and severity. The number of post-COVID symptoms was a significant predictor of PCD presence, while the number of acute symptoms was a significant predictor of PCD severity. Conclusions: This study, for the first time, reveals extensive demyelination in numerous WM and GM structures in PCD, outlining IFOF demyelination as a key biomarker.
Collapse
Affiliation(s)
- Marina Khodanovich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Mikhail Svetlik
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Daria Kamaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Anna Usova
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 12/1 Savinykh Street, Tomsk 634028, Russia
| | - Marina Kudabaeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Tatyana Anan’ina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Irina Vasserlauf
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Valentina Pashkevich
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Marina Moshkina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Victoria Obukhovskaya
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Department of Fundamental Psychology and Behavioral Medicine, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634050, Russia
| | - Nadezhda Kataeva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Department of Neurology and Neurosurgery, Siberian State Medical University, 2 Moskovskiy Trakt, Tomsk 634028, Russia
| | - Anastasia Levina
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
- Medica Diagnostic and Treatment Center, 86 Sovetskaya Street, Tomsk 634510, Russia
| | - Yana Tumentceva
- Laboratory of Neurobiology, Research Institute of Biology and Biophysics, Tomsk State University, 36 Lenina Ave., Tomsk 634050, Russia
| | - Svetlana Vasilieva
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Evgeny Schastnyy
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk 634014, Russia
| | - Anna Naumova
- Department of Radiology, School of Medicine, South Lake Union Campus, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| |
Collapse
|
12
|
Rodrigues-Amorim D, Bozzelli PL, Kim T, Liu L, Gibson O, Yang CY, Murdock MH, Galiana-Melendez F, Schatz B, Davison A, Islam MR, Shin Park D, Raju RM, Abdurrob F, Nelson AJ, Min Ren J, Yang V, Stokes MP, Tsai LH. Multisensory gamma stimulation mitigates the effects of demyelination induced by cuprizone in male mice. Nat Commun 2024; 15:6744. [PMID: 39112447 PMCID: PMC11306744 DOI: 10.1038/s41467-024-51003-7] [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: 09/02/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Demyelination is a common pathological feature in a wide range of diseases, characterized by the loss of myelin sheath and myelin-supporting oligodendrocytes. These losses lead to impaired axonal function, increased vulnerability of axons to damage, and result in significant brain atrophy and neuro-axonal degeneration. Multiple pathomolecular processes contribute to neuroinflammation, oligodendrocyte cell death, and progressive neuronal dysfunction. In this study, we use the cuprizone mouse model of demyelination to investigate long-term non-invasive gamma entrainment using sensory stimulation as a potential therapeutic intervention for promoting myelination and reducing neuroinflammation in male mice. Here, we show that multisensory gamma stimulation mitigates demyelination, promotes oligodendrogenesis, preserves functional integrity and synaptic plasticity, attenuates oligodendrocyte ferroptosis-induced cell death, and reduces brain inflammation. Thus, the protective effects of multisensory gamma stimulation on myelin and anti-neuroinflammatory properties support its potential as a therapeutic approach for demyelinating disorders.
Collapse
Grants
- R01 AG069232 NIA NIH HHS
- R01 AT011460 NCCIH NIH HHS
- R01 NS122742 NINDS NIH HHS
- R56 AG069232 NIA NIH HHS
- We would like to acknowledge the following individuals and organizations for their support: Fundacion Bancaria la Caixa, The JPB Foundation, Carol and Gene Ludwig Family Foundation, Lester A. Gimpelson, Eduardo Eurnekian, The Dolby Family, Kathy and Miguel Octavio, the Marc Haas Foundation, Ben Lenail and Laurie Yoler, and NIH RO1 grants AG069232, AT011460 and R01NS122742 to L.-H.T.
Collapse
Affiliation(s)
- Daniela Rodrigues-Amorim
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - P Lorenzo Bozzelli
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - TaeHyun Kim
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liwang Liu
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Cheng-Yi Yang
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mitchell H Murdock
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fabiola Galiana-Melendez
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brooke Schatz
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexis Davison
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Md Rezaul Islam
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dong Shin Park
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ravikiran M Raju
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Newborn Medicine, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Fatema Abdurrob
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Jian Min Ren
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA, USA
| | - Vicky Yang
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA, USA
| | | | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
13
|
Shen X, Caverzasi E, Yang Y, Liu X, Green A, Henry RG, Emir U, Larson PEZ. 3D balanced SSFP UTE MRI for multiple contrasts whole brain imaging. Magn Reson Med 2024; 92:702-714. [PMID: 38525680 DOI: 10.1002/mrm.30093] [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: 08/18/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE This study aimed to develop a new high-resolution MRI sequence for the imaging of the ultra-short transverse relaxation time (uT2) components in the brain, while simultaneously providing proton density (PD) contrast for reference and quantification. THEORY The sequence combines low flip angle balanced SSFP (bSSFP) and UTE techniques, together with a 3D dual-echo rosette k-space trajectory for readout. METHODS The expected image contrast was evaluated by simulations. A study cohort of six healthy volunteers and eight multiple sclerosis (MS) patients was recruited to test the proposed sequence. Subtraction between two TEs was performed to extract uT2 signals. In addition, conventional longitudinal relaxation time (T1) weighted, T2-weighted, and PD-weighted MRI sequences were also acquired for comparison. RESULTS Typical PD-contrast was found in the second TE images, while uT2 signals were selectively captured in the first TE images. The subtraction images presented signals primarily originating from uT2 components, but only if the first TE is short enough. Lesions in the MS subjects showed hyperintense signals in the second TE images but were hypointense signals in the subtraction images. The lesions had significantly lower signal intensity in subtraction images than normal white matter (WM), which indicated a reduction of uT2 components likely associated with myelin. CONCLUSION 3D isotropic sub-millimeter (0.94 mm) spatial resolution images were acquired with the novel bSSFP UTE sequence within 3 min. It provided easy extraction of uT2 signals and PD-contrast for reference within a single acquisition.
Collapse
Affiliation(s)
- Xin Shen
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Eduardo Caverzasi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Yang Yang
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Xiaoxi Liu
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Ari Green
- Neurology, University of California San Francisco, San Francisco, California, USA
| | - Roland G Henry
- Neurology, University of California San Francisco, San Francisco, California, USA
| | - Uzay Emir
- School of Health Science, Purdue University, West Lafayette, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Peder E Z Larson
- Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Sandgaard AD, Shemesh N, Østergaard L, Kiselev VG, Jespersen SN. The Larmor frequency shift of a white matter magnetic microstructure model with multiple sources. NMR IN BIOMEDICINE 2024; 37:e5150. [PMID: 38553824 DOI: 10.1002/nbm.5150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 07/11/2024]
Abstract
Magnetic susceptibility imaging may provide valuable information about chemical composition and microstructural organization of tissue. However, its estimation from the MRI signal phase is particularly difficult as it is sensitive to magnetic tissue properties ranging from the molecular to the macroscopic scale. The MRI Larmor frequency shift measured in white matter (WM) tissue depends on the myelinated axons and other magnetizable sources such as iron-filled ferritin. We have previously derived the Larmor frequency shift arising from a dense medium of cylinders with scalar susceptibility and arbitrary orientation dispersion. Here, we extend our model to include microscopic WM susceptibility anisotropy as well as spherical inclusions with scalar susceptibility to represent subcellular structures, biologically stored iron, and so forth. We validate our analytical results with computer simulations and investigate the feasibility of estimating susceptibility using simple iterative linear least squares without regularization or preconditioning. This is done in a digital brain phantom synthesized from diffusion MRI measurements of an ex vivo mouse brain at ultra-high field.
Collapse
Affiliation(s)
- Anders Dyhr Sandgaard
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Valerij G Kiselev
- Division of Medical Physics, Department of Radiology, University Medical Center Freiburg, Freiburg, Germany
| | - Sune Nørhøj Jespersen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Physics and Astronomy, Aarhus University, Aarhus, Denmark
| |
Collapse
|
15
|
Moghimyfiroozabad S, Paul MA, Bellenger L, Selimi F. A molecularly defined subpopulation of oligodendrocyte precursor cells controls the generation of myelinating oligodendrocytes during postnatal development. PLoS Biol 2024; 22:e3002655. [PMID: 38985832 PMCID: PMC11236193 DOI: 10.1371/journal.pbio.3002655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 05/02/2024] [Indexed: 07/12/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are a class of glial cells that uniformly tiles the entire central nervous system (CNS). They play several key functions across the brain including the generation of oligodendrocytes and the control of myelination. Whether the functional diversity of OPCs is the result of genetically defined subpopulations or of their regulation by external factors has not been definitely established. We discovered that a subpopulation of OPCs found across the brain is defined by the expression of C1ql1, a gene previously described for its synaptic function in neurons. This subpopulation starts to appear during the first postnatal week in the mouse cortex. Ablation of C1ql1-expressing OPCs in the mouse leads to a massive lack of oligodendrocytes and myelination in many brain regions. This deficit cannot be rescued, even though some OPCs escape Sox10-driven ablation and end up partially compensating the OPC loss in the adult. Therefore, C1ql1 is a molecular marker of a functionally non-redundant subpopulation of OPCs, which controls the generation of myelinating oligodendrocytes.
Collapse
Affiliation(s)
- Shayan Moghimyfiroozabad
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Maela A Paul
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Lea Bellenger
- ARTbio Bioinformatics Analysis Facility, Sorbonne Université, Inserm U1156, CNRS FR 3631, Institut Français de Bioinformatique (IFB), Paris, France
| | - Fekrije Selimi
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
16
|
Affrald R J, Narayan S. A review: oligodendrocytes in neuronal axonal conduction and methods for enhancing their performance. Int J Neurosci 2024:1-22. [PMID: 38850232 DOI: 10.1080/00207454.2024.2362200] [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: 07/06/2022] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVES This review explores the vital role of oligodendrocytes in axon myelination and efficient neuronal transmission and the impact of dysfunction resulting from neurotransmitter deficiencies related disorders. Furthermore, the review also provides insight into the potential of bionanotechnology for addressing neurodegenerative diseases by targeting oligodendrocytes. METHODS A review of literature in the field was conducted using Google scholar. Systematic searches were performed to identify relevant studies and reviews addressing the role of oligodendrocytes in neural function, the influence of neurotransmitters on oligodendrocyte differentiation, and the potential of nanotechnology-based strategies for targeted therapy of oligodendrocytes. RESULTS This review indicates the mechanisms underlying oligodendrocyte differentiation and the influence of neurotransmitters on this process. The importance of action potentials and neurotransmission in neural function and the susceptibility of damaged nerve axons to ischemic or toxic damage is provided in detail. The potential of bionanotechnology for targeting neurodegenerative diseases using nanotechnology-based strategies, including polymeric, lipid-based, inorganic, organic, and biomimetic nanoparticles, suggests better management of neurodegenerative disorders. CONCLUSION While nanotechnology-based biomaterials show promise for targeted oligodendrocyte therapy in addressing neurodegenerative disorders linked to oligodendrocyte dysfunction, encapsulating neuroprotective agents within nanoparticles offers additional advantages. Nano-based delivery systems effectively protect drugs from degradation and prolong their therapeutic effects, holding promise in overcoming the blood-brain barrier by facilitating drug transport. However, a multifaceted approach is essential to enhance oligodendrocyte differentiation, promote myelin repair, and facilitate myelin dynamics with reduced toxicity. Further research is needed to elucidate the optimal therapeutic approaches and enhance patient outcomes.
Collapse
Affiliation(s)
- Jino Affrald R
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, India
| | - Shoba Narayan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamilnadu, India
| |
Collapse
|
17
|
Kawaguchi S, Kan H, Uchida Y, Kasai H, Hiwatashi A, Ueki Y. Anisotropy of the R1/T2* value dependent on white matter fiber orientation with respect to the B0 field. Magn Reson Imaging 2024; 109:83-90. [PMID: 38387713 DOI: 10.1016/j.mri.2024.02.010] [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: 12/26/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
The R1 (1/T1) map divided by the T2* map (R1/T2* map) draws attention as a high-resolution myelin-related map. However, both R1 and R2* (1/T2*) values demonstrate anisotropy dependent on the white matter (WM) fiber orientation with respect to the static magnetic (B0) field. Therefore, this study primarily aimed to investigate the comprehensive impact of these angular-dependent anisotropies on the R1/T2* value. This study enrolled 10 healthy human volunteers (age = 25 ± 1.3) on the 3.0 T MRI system. For R1/T2* map calculation, whole brain R1 and T2* maps were repeatedly obtained in three head tilt positions by magnetization-prepared two rapid gradient echoes and multiple spoiled gradient echo sequences, respectively. Afterward, all maps were spatially normalized and registered to the Johns Hopkins University WM atlas. R1/T2*, R1, and R2* values were binned for fiber orientation related to the B0 field, which was estimated from diffusion-weighted echo-planar imaging data with 3° intervals, to investigate angular-dependent anisotropies in vivo. A larger change in the R1/T2* value in the global WM region as a function of fiber orientation with respect to the B0 field was observed compared to the R1 and R2* values alone. The minimum R1/T2* value at the near magic-angle range was 18.86% lower than the maximum value at the perpendicular angle range. Furthermore, R1/T2* values in the corpus callosum tract and the right and left cingulum cingulate gyrus tracts changed among the three head tilt positions due to fiber orientation changes. In conclusion, the R1/T2* value demonstrates distinctive and complicated angular-dependent anisotropy indicating the trends of both R1 and R2* values and may provide supplemental information for detecting slight changes in the microstructure of myelin and axons.
Collapse
Affiliation(s)
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Japan; Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Japan.
| | - Yuto Uchida
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Harumasa Kasai
- Department of Radiology, Nagoya City University Hospital, Japan
| | - Akio Hiwatashi
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Yoshino Ueki
- Department of Rehabilitation Medicine, Nagoya City University Graduate School of Medical Sciences, Japan
| |
Collapse
|
18
|
Gharighnia S, Omidi A, Ragerdi Kashani I, Sepand MR, Pour Beiranvand S. Ameliorative effects of acetyl-L-carnitine on corpus callosum and functional recovery in demyelinated mouse model. Int J Neurosci 2024; 134:409-419. [PMID: 35912879 DOI: 10.1080/00207454.2022.2107515] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
AIM Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disease of the central nervous system. Oxidative stress via distinct pathobiological pathways plays a pivotal role in the formation and persistence of MS lesions. Acetyl-L-carnitine (ALC) facilitates the uptake of acetyl coenzyme-A into the mitochondria by a fatty acid oxidation process. ALC could be a therapeutic antioxidant in the myelin repair process. This study explored the potential neuroprotective effects of ALC in cuprizone (CPZ) intoxicated mice. MATERIALS AND METHODS Thirty male C57BL/6 mice were divided into three groups. The control animals received a normal diet. The CPZ and CPZ + ALC groups were fed with a 0.2% cuprizone diet for 12 weeks. In the CPZ + ALC group, animals received ALC (300 mg/kg/day) from the 10th -12th weeks. Animals were evaluated functionally by beam walking test (BWT) weekly. Eventually, the corpus callosum (CC) was extracted for histological, biochemical, and molecular studies. RESULTS BWT data showed ALC significantly improves balance and gait in the demyelinating mouse model. Histological staining represented ALC effectively increased remyelination in the CC. Biochemical evaluations demonstrated ALC decreased the malondialdehyde level with a parallel increase in the reduced glutathione and catalase activity levels in the CC. Molecular analysis revealed that ALC significantly increased the expression of oligodendrocyte transcription-2 (Olig-2) and Poly lipoproteins (Plp) genes in the CC. CONCLUSIONS ALC improved balance and motor coordination in the demyelinated mouse model. It may be by reducing the levels of free radicals and increasing the expression of Olig-2 and Plp as myelin-related genes.
Collapse
Affiliation(s)
- Sanaz Gharighnia
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Sepand
- Toxicology and Poisoning Research Centre, Department of Toxicology and Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Pour Beiranvand
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
19
|
de la Fuente AG, Dittmer M, Heesbeen EJ, de la Vega Gallardo N, White JA, Young A, McColgan T, Dashwood A, Mayne K, Cabeza-Fernández S, Falconer J, Rodriguez-Baena FJ, McMurran CE, Inayatullah M, Rawji KS, Franklin RJM, Dooley J, Liston A, Ingram RJ, Tiwari VK, Penalva R, Dombrowski Y, Fitzgerald DC. Ageing impairs the regenerative capacity of regulatory T cells in mouse central nervous system remyelination. Nat Commun 2024; 15:1870. [PMID: 38467607 PMCID: PMC10928230 DOI: 10.1038/s41467-024-45742-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: 01/15/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Myelin regeneration (remyelination) is essential to prevent neurodegeneration in demyelinating diseases such as Multiple Sclerosis, however, its efficiency declines with age. Regulatory T cells (Treg) recently emerged as critical players in tissue regeneration, including remyelination. However, the effect of ageing on Treg-mediated regenerative processes is poorly understood. Here, we show that expansion of aged Treg does not rescue age-associated remyelination impairment due to an intrinsically diminished capacity of aged Treg to promote oligodendrocyte differentiation and myelination in male and female mice. This decline in regenerative Treg functions can be rescued by a young environment. We identified Melanoma Cell Adhesion Molecule 1 (MCAM1) and Integrin alpha 2 (ITGA2) as candidates of Treg-mediated oligodendrocyte differentiation that decrease with age. Our findings demonstrate that ageing limits the neuroregenerative capacity of Treg, likely limiting their remyelinating therapeutic potential in aged patients, and describe two mechanisms implicated in Treg-driven remyelination that may be targetable to overcome this limitation.
Collapse
Affiliation(s)
- Alerie Guzman de la Fuente
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
- Institute for Health and Biomedical Sciences of Alicante (ISABIAL), Alicante, 03010, Spain.
- Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Alicante, 03550, Spain.
| | - Marie Dittmer
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Elise J Heesbeen
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Nira de la Vega Gallardo
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Jessica A White
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Andrew Young
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Tiree McColgan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Amy Dashwood
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
- Babraham Institute, CB22 3AT, Cambridge, UK
| | - Katie Mayne
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Sonia Cabeza-Fernández
- Institute for Health and Biomedical Sciences of Alicante (ISABIAL), Alicante, 03010, Spain
- Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Alicante, 03550, Spain
| | - John Falconer
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- CRUK Beatson Institute, G61 1BD, Glasgow, UK
| | | | - Christopher E McMurran
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Mohammed Inayatullah
- Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Danish Institute for Advanced Study (DIAS), 5230, Odense, Denmark
| | - Khalil S Rawji
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge, CB21 6GP, UK
| | - Robin J M Franklin
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge, CB21 6GP, UK
| | - James Dooley
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
| | - Adrian Liston
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Danish Institute for Advanced Study (DIAS), 5230, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, 5000, Odense, Denmark
| | - Rosana Penalva
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Yvonne Dombrowski
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Denise C Fitzgerald
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
20
|
El Homsi M, Zadeh C, Charbel C, Alsheikh Deeb I, Gharzeddine K, Rebeiz K, Hourani R, Khoury N, Moukaddam H. Neurologic pathologies of the vertebral spine. Skeletal Radiol 2024; 53:419-436. [PMID: 37589755 DOI: 10.1007/s00256-023-04428-y] [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: 03/14/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023]
Abstract
At some institutions, musculoskeletal and general radiologists rather than neuroradiologists are responsible for reading magnetic resonance imaging (MRI) of the spine. However, neurological findings, especially intrathecal ones, can be challenging. Intrathecal neurological findings in the spine can be classified by location (epidural, intradural extramedullary, and intramedullary) or etiology (tumor, infection, inflammatory, congenital). In this paper, we provide a succinct review of the intrathecal neurological findings that can be seen on MRI of the spine, primarily by location and secondarily by etiology, in order that this may serve as a helpful guide for musculoskeletal and general radiologists when encountering intrathecal neurological pathologies.
Collapse
Affiliation(s)
- Maria El Homsi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Catherina Zadeh
- Department of Radiology, University of Iowa Hospital and Clinics, Iowa, IA, USA
| | - Charlotte Charbel
- Department of Radiology, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Ibrahim Alsheikh Deeb
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Karem Gharzeddine
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karim Rebeiz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roula Hourani
- Department of Diagnostic Radiology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nabil Khoury
- Department of Radiology, University of Iowa Hospital and Clinics, Iowa, IA, USA
- Department of Diagnostic Radiology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hicham Moukaddam
- Department of Diagnostic Radiology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
21
|
Tejwani L, Ravindra NG, Lee C, Cheng Y, Nguyen B, Luttik K, Ni L, Zhang S, Morrison LM, Gionco J, Xiang Y, Yoon J, Ro H, Haidery F, Grijalva RM, Bae E, Kim K, Martuscello RT, Orr HT, Zoghbi HY, McLoughlin HS, Ranum LPW, Shakkottai VG, Faust PL, Wang S, van Dijk D, Lim J. Longitudinal single-cell transcriptional dynamics throughout neurodegeneration in SCA1. Neuron 2024; 112:362-383.e15. [PMID: 38016472 PMCID: PMC10922326 DOI: 10.1016/j.neuron.2023.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 09/10/2023] [Accepted: 10/27/2023] [Indexed: 11/30/2023]
Abstract
Neurodegeneration is a protracted process involving progressive changes in myriad cell types that ultimately results in the death of vulnerable neuronal populations. To dissect how individual cell types within a heterogeneous tissue contribute to the pathogenesis and progression of a neurodegenerative disorder, we performed longitudinal single-nucleus RNA sequencing of mouse and human spinocerebellar ataxia type 1 (SCA1) cerebellar tissue, establishing continuous dynamic trajectories of each cell population. Importantly, we defined the precise transcriptional changes that precede loss of Purkinje cells and, for the first time, identified robust early transcriptional dysregulation in unipolar brush cells and oligodendroglia. Finally, we applied a deep learning method to predict disease state accurately and identified specific features that enable accurate distinction of wild-type and SCA1 cells. Together, this work reveals new roles for diverse cerebellar cell types in SCA1 and provides a generalizable analysis framework for studying neurodegeneration.
Collapse
Affiliation(s)
- Leon Tejwani
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Neal G Ravindra
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Computer Science, Yale University, New Haven, CT 06510, USA
| | - Changwoo Lee
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yubao Cheng
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Billy Nguyen
- University of California, San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Kimberly Luttik
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Luhan Ni
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shupei Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Logan M Morrison
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - John Gionco
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY 10032, USA
| | - Yangfei Xiang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Hannah Ro
- Yale College, New Haven, CT 06510, USA
| | | | - Rosalie M Grijalva
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Kristen Kim
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA
| | - Regina T Martuscello
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY 10032, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hayley S McLoughlin
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Laura P W Ranum
- Department of Molecular Genetics and Microbiology, Center for Neurogenetics, College of Medicine, Genetics Institute, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Phyllis L Faust
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY 10032, USA
| | - Siyuan Wang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA.
| | - David van Dijk
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Computer Science, Yale University, New Haven, CT 06510, USA.
| | - Janghoo Lim
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA; Wu Tsai Institute, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
22
|
Mousavi SH, Lindsey JW, Westlund KN, Alles SRA. Trigeminal Neuralgia as a Primary Demyelinating Disease: Potential Multimodal Evidence and Remaining Controversies. THE JOURNAL OF PAIN 2024; 25:302-311. [PMID: 37643657 DOI: 10.1016/j.jpain.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Trigeminal neuralgia is a heterogeneous disorder with likely multifactorial and complex etiology; however, trigeminal nerve demyelination and injury are observed in almost all patients with trigeminal neuralgia. The current management strategies for trigeminal neuralgia primarily involve anticonvulsants and surgical interventions, neither of which directly address demyelination, the pathological hallmark of trigeminal neuralgia, and treatments targeting demyelination are not available. Demyelination of the trigeminal nerve has been historically considered a secondary effect of vascular compression, and as a result, trigeminal neuralgia is not recognized nor treated as a primary demyelinating disorder. In this article, we review the evolution of our understanding of trigeminal neuralgia and provide evidence to propose its potential categorization, at least in some cases, as a primary demyelinating disease by discussing its course and similarities to multiple sclerosis, the most prevalent central nervous system demyelinating disorder. This proposed categorization may provide a basis in investigating novel treatment modalities beyond the current medical and surgical interventions, emphasizing the need for further research into demyelination of the trigeminal sensory pathway in trigeminal neuralgia. PERSPECTIVE: This article proposes trigeminal neuralgia as a demyelinating disease, supported by histological, clinical, and radiological evidence. Such categorization offers a plausible explanation for controversies surrounding trigeminal neuralgia. This perspective holds potential for future research and developing therapeutics targeting demyelination in the condition.
Collapse
Affiliation(s)
- Seyed H Mousavi
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas
| | - John W Lindsey
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, University of Texas Health Science Center at Houston (UTHealth), Houston, Texas
| | - Karin N Westlund
- Department of Anesthesiology & Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sascha R A Alles
- Department of Anesthesiology & Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
23
|
Chen HH, Lercara C, Lee V, Bushi S. Rehabilitation after Hypoxic and Metabolic Brain Injury in a Mountain Climber. BMJ Case Rep 2024; 17:e255794. [PMID: 38238166 PMCID: PMC10806977 DOI: 10.1136/bcr-2023-255794] [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] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
A patient in her 50s presented with altered mental status and shortness of breath at 4600 m elevation. After descent to the base of the mountain, the patient became comatose. She was found to have bilateral pulmonary infiltrates and a serum sodium of 102 mEq/L. She was rapidly corrected to 131 mEq/L in 1 day. Initial MRI showed intensities in bilateral hippocampi, temporal cortex and insula. A repeat MRI 17 days post injury showed worsened intensities in the bilateral occipital lobes. On admission to acute rehabilitation, the patient presented with blindness, agitation, hallucinations and an inability to follow commands. Midway through her rehabilitation course, antioxidant supplementations were started with significant improvement in function. Rapid correction of hyponatraemia may cause central pontine myelinolysis or extrapontine myelinolysis (EPM). In some cases of hypoxic brain injury, delayed post-hypoxic leucoencephalopathy (DPHL) may occur. Treatment options for both disorders are generally supportive. This report represents the only documented interdisciplinary approach to treatment of a patient with DPHL and EPM. Antioxidant supplementation may be beneficial as a treatment option for both EPM and DPHL.
Collapse
Affiliation(s)
- Henry Han Chen
- Department of Physical Medicine and Rehabilitation, Burke Rehabilitation Hospital, White Plains, New York, USA
| | | | - Vincent Lee
- Department of Physical Medicine and Rehabilitation, Burke Rehabilitation Hospital, White Plains, New York, USA
| | - Sharon Bushi
- Department of Physical Medicine and Rehabilitation, Burke Rehabilitation Hospital, White Plains, New York, USA
- Montefiore Health System, Bronx, New York, USA
| |
Collapse
|
24
|
Zhou D, Xu L, Wang T, Wei S, Gao F, Lai X, Cao J. M-DDC: MRI based demyelinative diseases classification with U-Net segmentation and convolutional network. Neural Netw 2024; 169:108-119. [PMID: 37890361 DOI: 10.1016/j.neunet.2023.10.010] [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: 12/26/2022] [Revised: 09/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
Childhood demyelinative diseases classification (DDC) with brain magnetic resonance imaging (MRI) is crucial to clinical diagnosis. But few attentions have been paid to DDC in the past. How to accurately differentiate pediatric-onset neuromyelitis optica spectrum disorder (NMOSD) from acute disseminated encephalomyelitis (ADEM) based on MRI is challenging in DDC. In this paper, a novel architecture M-DDC based on joint U-Net segmentation network and deep convolutional network is developed. The U-Net segmentation can provide pixel-level structure information, that helps the lesion areas location and size estimation. The classification branch in DDC can detect the regions of interest inside MRIs, including the white matter regions where lesions appear. The performance of the proposed method is evaluated on MRIs of 201 subjects recorded from the Children's Hospital of Zhejiang University School of Medicine. The comparisons show that the proposed DDC achieves the highest accuracy of 99.19% and dice of 71.1% for ADEM and NMOSD classification and segmentation, respectively.
Collapse
Affiliation(s)
- Deyang Zhou
- Machine Learning and I-health International Cooperation Base of Zhejiang Province, Hangzhou Dianzi University, 310018, China; Artificial Intelligence Institute, Hangzhou Dianzi University, Zhejiang, 310018, China; HDU-ITMO Joint Institute, Hangzhou Dianzi University, Zhejiang, 310018, China.
| | - Lu Xu
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, 310018, China.
| | - Tianlei Wang
- Machine Learning and I-health International Cooperation Base of Zhejiang Province, Hangzhou Dianzi University, 310018, China; Artificial Intelligence Institute, Hangzhou Dianzi University, Zhejiang, 310018, China.
| | - Shaonong Wei
- Machine Learning and I-health International Cooperation Base of Zhejiang Province, Hangzhou Dianzi University, 310018, China; Artificial Intelligence Institute, Hangzhou Dianzi University, Zhejiang, 310018, China; HDU-ITMO Joint Institute, Hangzhou Dianzi University, Zhejiang, 310018, China.
| | - Feng Gao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, 310018, China.
| | - Xiaoping Lai
- Machine Learning and I-health International Cooperation Base of Zhejiang Province, Hangzhou Dianzi University, 310018, China; Artificial Intelligence Institute, Hangzhou Dianzi University, Zhejiang, 310018, China.
| | - Jiuwen Cao
- Machine Learning and I-health International Cooperation Base of Zhejiang Province, Hangzhou Dianzi University, 310018, China; Artificial Intelligence Institute, Hangzhou Dianzi University, Zhejiang, 310018, China.
| |
Collapse
|
25
|
Gakare SG, Bhatt JM, Narasimhan KKS, Dravid SM. Glutamate delta-1 receptor regulates oligodendrocyte progenitor cell differentiation and myelination in normal and demyelinating conditions. PLoS One 2023; 18:e0294583. [PMID: 37983226 PMCID: PMC10659214 DOI: 10.1371/journal.pone.0294583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
In this study, we investigated the role of glutamate delta 1 receptor (GluD1) in oligodendrocyte progenitor cell (OPC)-mediated myelination during basal (development) and pathophysiological (cuprizone-induced demyelination) conditions. Initially, we sought to determine the expression pattern of GluD1 in OPCs and found a significant colocalization of GluD1 puncta with neuron-glial antigen 2 (NG2, OPC marker) in the motor cortex and dorsal striatum. Importantly, we found that the ablation of GluD1 led to an increase in the number of myelin-associated glycoprotein (MAG+) cells in the corpus callosum and motor cortex at P40 without affecting the number of NG2+ OPCs, suggesting that GluD1 loss selectively facilitates OPC differentiation rather than proliferation. Further, deletion of GluD1 enhanced myelination in the corpus callosum and motor cortex, as indicated by increased myelin basic protein (MBP) staining at P40, suggesting that GluD1 may play an essential role in the developmental regulation of myelination during the critical window period. In contrast, in cuprizone-induced demyelination, we observed reduced MBP staining in the corpus callosum of GluD1 KO mice. Furthermore, cuprizone-fed GluD1 KO mice showed more robust motor deficits. Collectively, our results demonstrate that GluD1 plays a critical role in OPC regulation and myelination in normal and demyelinating conditions.
Collapse
Affiliation(s)
- Sukanya G. Gakare
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Jay M. Bhatt
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Kishore Kumar S. Narasimhan
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Shashank M. Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| |
Collapse
|
26
|
Jagielska A, Radzwill K, Espinosa-Hoyos D, Yang M, Kowsari K, Farley JE, Giera S, Byrne A, Sheng G, Fang NX, Dodge JC, Pedraza CE, Van Vliet KJ. Artificial axons as a biomimetic 3D myelination platform for the discovery and validation of promyelinating compounds. Sci Rep 2023; 13:19529. [PMID: 37945646 PMCID: PMC10636046 DOI: 10.1038/s41598-023-44675-6] [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: 02/24/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023] Open
Abstract
Multiple sclerosis (MS), a chronic neurodegenerative disease driven by damage to the protective myelin sheath, is currently incurable. Today, all clinically available treatments modulate the immune-mediated symptoms of the disease but they fail to stop neurodegeneration in many patients. Remyelination, the regenerative process of myelin repair by oligodendrocytes, which is considered a necessary step to protect demyelinated axons and stop neuronal death, is impaired in MS patients. One of the major obstacles to finding effective remyelinating drugs is the lack of biomimetic drug screening platforms that enable quantification of compounds' potential to stimulate 3D myelination in the physiologically relevant axon-like environment. To address this need, we built a unique myelination drug discovery platform, by expanding our previously developed technology, artificial axons (AAs), which enables 3D-printing of synthetic axon mimics with the geometry and mechanical properties closely resembling those of biological axons. This platform allows for high-throughput phenotypic myelination assay based on quantification of 3D wrapping of myelin membrane around axons in response to compounds. Here, we demonstrate quantification of 3D myelin wrapping by rat oligodendrocytes around the axon mimics in response to a small library of known pro-myelinating compounds. This assay shows pro-myelinating activity for all tested compounds consistent with the published in vitro and in vivo data, demonstrating predictive power of AA platform. We find that stimulation of myelin wrapping by these compounds is dose-dependent, providing a facile means to quantify the compounds' potency and efficacy in promoting myelin wrapping. Further, the ranking of relative efficacy among these compounds differs in this 3D axon-like environment as compared to a traditional oligodendrocyte 2D differentiation assay quantifying area of deposited myelin membrane. Together, we demonstrate that the artificial axons platform and associated phenotypic myelin wrapping assay afford direct evaluation of myelin wrapping by oligodendrocytes in response to soluble compounds in an axon-like environment, providing a predictive tool for the discovery of remyelinating therapies.
Collapse
Affiliation(s)
- Anna Jagielska
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Artificial Axon Labs, Boston, MA, USA.
| | | | - Daniela Espinosa-Hoyos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Sanofi, Cambridge, MA, USA
| | - Mingyu Yang
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kavin Kowsari
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Merck, Rahway, NJ, USA
| | - Jonathan E Farley
- Sanofi, Cambridge, MA, USA
- Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | - Nicholas X Fang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The University of Hong Kong, Hong Kong, China
| | | | | | - Krystyn J Van Vliet
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Cornell University, Ithaca, NY, USA.
| |
Collapse
|
27
|
Fazlollahi A, Zahmatyar M, Shamekh A, Motamedi A, Seyedi F, Seyedmirzaei H, Mousavi SE, Nejadghaderi SA, Sullman MJM, Kolahi AA, Arshi S, Safiri S. Electroencephalographic findings post-COVID-19 vaccination: A systematic review of case reports and case series. Rev Med Virol 2023; 33:e2484. [PMID: 37807809 DOI: 10.1002/rmv.2484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/14/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
A number of different neurological complications have been reported following vaccination against the coronavirus disease 2019 (COVID-19). Electroencephalogram (EEG) is one of the modalities used to evaluate the neurological complications of diseases. The aim of the present study was to identify the EEG changes in participants vaccinated against COVID-19. PubMed, Scopus, Web of Science, medRxiv, and Google Scholar were searched up to 1 September 2022, with terms related to COVID-19 vaccines, EEG, neurological signs/symptoms, or neurological disorders. All case reports and case series were included if the participants had received at least one dose of a COVID-19 vaccine and a post vaccination EEG report was also reported. We used the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for case reports and case series to appraise the methodological quality of the included studies. Thirty-one studies were included, which were comprised of 24 case reports and seven case series and a total of 36 participants. Generalised slowing and non-convulsive focal status epilepticus were the most common EEG findings post-COVID-19 vaccination. The most frequent symptoms were headache, fatigue, generalised weakness, and vomiting. In addition, the most common signs were encephalopathy, post-ictal phases, and confusion. Encephalitis, acute disseminated encephalomyelitis, and post-vaccinal encephalopathy were the most commonly diagnosed adverse events. Furthermore, most of the imaging studies appeared normal. The EEG reports mainly showed background slowing and epileptiform discharges, encephalitis, encephalopathies, and demyelinating disorders. Future studies with larger samples and more vaccine types may help to further unravel the potential neurological effects of COVID-19 vaccinations on recipients.
Collapse
Affiliation(s)
- Asra Fazlollahi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Zahmatyar
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Motamedi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Seyedi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and Lung Diseases Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Aria Nejadghaderi
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahnam Arshi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Balloff C, Novello S, Stucke AS, Janssen LK, Heinen E, Hartmann CJ, Meuth SG, Schnitzler A, Penner IK, Albrecht P, Groiss SJ. Long-term potentiation-like plasticity is retained during relapse in patients with Multiple Sclerosis. Clin Neurophysiol 2023; 155:76-85. [PMID: 37776674 DOI: 10.1016/j.clinph.2023.07.013] [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: 03/04/2023] [Revised: 06/23/2023] [Accepted: 07/19/2023] [Indexed: 10/02/2023]
Abstract
OBJECTIVE To investigate the degree of synaptic plasticity in Multiple Sclerosis (MS) patients during acute relapses compared to stable MS patients and healthy controls (HCs) and to analyze its functional relevance. METHODS Facilitatory quadripulse stimulation (QPS) was applied to the primary motor cortex in 18 acute relapsing and 18 stable MS patients, as well as 18 HCs. The degree of synaptic plasticity was measured by the change in motor evoked potential amplitude following QPS. Symptom recovery was assessed three months after relapse. RESULTS Synaptic plasticity was induced in all groups. The degree of induced plasticity did not differ between acute relapsing patients, HCs, and stable MS patients. Plasticity was significantly higher in relapsing patients with motor disability compared to relapsing patients without motor disability. In most patients (n = 9, 50%) symptoms had at least partially recovered three months after the relapse, impeding meaningful analysis of the functional relevance of baseline synaptic plasticity. CONCLUSIONS QPS-induced synaptic plasticity is retained during acute MS relapses. Subgroup analyses suggest that stabilizing metaplastic mechanisms may be more important to prevent motor disability but its functional relevance needs to be verified in larger, longitudinal studies. SIGNIFICANCE New insights into synaptic plasticity during MS relapses are provided.
Collapse
Affiliation(s)
- Carolin Balloff
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Department of Neurology, Kliniken Maria Hilf GmbH, 41063 Moenchengladbach, Germany
| | - Sveva Novello
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Arved-Sebastian Stucke
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Lisa Kathleen Janssen
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Elisa Heinen
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Christian Johannes Hartmann
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Sven Günther Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Alfons Schnitzler
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Iris-Katharina Penner
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Department of Neurology, Kliniken Maria Hilf GmbH, 41063 Moenchengladbach, Germany.
| | - Stefan Jun Groiss
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany; Neurocenter Duesseldorf, 40211 Duesseldorf, Germany
| |
Collapse
|
29
|
Saksena J, Hamilton AE, Gilbert RJ, Zuidema JM. Nanomaterial payload delivery to central nervous system glia for neural protection and repair. Front Cell Neurosci 2023; 17:1266019. [PMID: 37941607 PMCID: PMC10628439 DOI: 10.3389/fncel.2023.1266019] [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: 07/24/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Central nervous system (CNS) glia, including astrocytes, microglia, and oligodendrocytes, play prominent roles in traumatic injury and degenerative disorders. Due to their importance, active pharmaceutical ingredients (APIs) are being developed to modulate CNS glia in order to improve outcomes in traumatic injury and disease. While many of these APIs show promise in vitro, the majority of APIs that are systemically delivered show little penetration through the blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB) and into the CNS, rendering them ineffective. Novel nanomaterials are being developed to deliver APIs into the CNS to modulate glial responses and improve outcomes in injury and disease. Nanomaterials are attractive options as therapies for central nervous system protection and repair in degenerative disorders and traumatic injury due to their intrinsic capabilities in API delivery. Nanomaterials can improve API accumulation in the CNS by increasing permeation through the BBB of systemically delivered APIs, extending the timeline of API release, and interacting biophysically with CNS cell populations due to their mechanical properties and nanoscale architectures. In this review, we present the recent advances in the fields of both locally implanted nanomaterials and systemically administered nanoparticles developed for the delivery of APIs to the CNS that modulate glial activity as a strategy to improve outcomes in traumatic injury and disease. We identify current research gaps and discuss potential developments in the field that will continue to translate the use of glia-targeting nanomaterials to the clinic.
Collapse
Affiliation(s)
- Jayant Saksena
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Adelle E. Hamilton
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Albany Stratton Veterans Affairs Medical Center, Albany, NY, United States
| | - Jonathan M. Zuidema
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| |
Collapse
|
30
|
Nagarajan A, Rizwana N, Abraham M, Bhat M, Vetekar A, Thakur G, Chakraborty U, Agarwal V, Nune M. Polycaprolactone/graphene oxide/acellular matrix nanofibrous scaffolds with antioxidant and promyelinating features for the treatment of peripheral demyelinating diseases. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:49. [PMID: 37796399 PMCID: PMC10556163 DOI: 10.1007/s10856-023-06750-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 08/24/2023] [Indexed: 10/06/2023]
Abstract
Peripheral demyelinating diseases entail damage to axons and Schwann cells in the peripheral nervous system. Because of poor prognosis and lack of a cure, this group of diseases has a global impact. The primary underlying cause of these diseases involves the inability of Schwann cells to remyelinate the damaged insulating myelin around axons, resulting in neuronal death over time. In the past decade, extensive research has been directed in the direction of Schwann cells focusing on their physiological and neuroprotective effects on the neurons in the peripheral nervous system. One cause of dysregulation in the remyelinating function of Schwann cells has been associated with oxidative stress. Tissue-engineered biodegradable scaffolds that can stimulate remyelination response in Schwann cells have been proposed as a potential treatment strategy for peripheral demyelinating diseases. However, strategies developed to date primarily focussed on either remyelination or oxidative stress in isolation. Here, we have developed a multifunctional nanofibrous scaffold with material and biochemical cues to tackle both remyelination and oxidative stress in one matrix. We developed a nanofibrous scaffold using polycaprolactone (PCL) as a foundation loaded with antioxidant graphene oxide (GO) and coated this bioscaffold with Schwann cell acellular matrix. In vitro studies revealed both antioxidant and remyelination properties of the developed bioscaffold. Based on the results, the developed multifunctional bioscaffold approach can be a promising biomaterial approach for treating demyelinating diseases.
Collapse
Affiliation(s)
- Aishwarya Nagarajan
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Nasera Rizwana
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Michelle Abraham
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mahima Bhat
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Aakanksha Vetekar
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
- Department. of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Goutam Thakur
- Department. of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Uttara Chakraborty
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Bengaluru, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
31
|
Al-Tameemi HN, Hassoun HK, Mohammed IQ, Allebban Z. MRI assessment of cervical spinal cord cross-sectional area in patients with multiple sclerosis. J Neurosci Rural Pract 2023; 14:660-666. [PMID: 38059247 PMCID: PMC10696324 DOI: 10.25259/jnrp_87_2023] [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: 02/23/2023] [Accepted: 06/30/2023] [Indexed: 12/08/2023] Open
Abstract
Objectives Spinal cord abnormalities including cervical cord atrophy are common in multiple sclerosis (MS). This study aimed to assess the cervical spinal cord cross-sectional area (CSA) using magnetic resonance imaging (MRI) in MS patients. Materials and Methods Sixty participants were enrolled in this study (16 male and 44 female), 30 patients with MS, diagnosed according to the revised McDonald criteria, and 30 apparently healthy individuals as the control group. CSA of the spinal cord was measured on axial T2-weighted images of the cervical MRI studies from C2 to C7 vertebral levels. Results There was a significant difference between MS patients and the control group in mean CSA at a different level. The mean CSA at C2, in MS cases, was significantly lower than controls (67.7 ± 9.4 mm2 vs. 81.3 ± 4.6 mm2). Similarly, the mean CSA at C7 (64.4 ± 9.9 mm2) and average C2-7 (68 ± 9.1 mm2) of MS cases were significantly lower than the control. There was a strong inverse correlation between mean cervical cord CSA and duration of the disease and disability score. The reduction in cervical cord CSA was more prominent in patients with secondary progressive MS. There was no significant difference regarding age, gender, type of treatment, or the number of cervical cord lesions. Conclusion The mean CSA was significantly lower in patients with MS than in the control group and was lesser in progressive types. Patients with a longer duration of MS and a high disability score tend to have smaller CSA.
Collapse
Affiliation(s)
- Haider N. Al-Tameemi
- Middle Euphrates Neurosciences Center, Faculty of Medicine, Kufa University, Al-Najaf, Iraq
| | - Hayder K. Hassoun
- Middle Euphrates Neurosciences Center, Faculty of Medicine, Kufa University, Al-Najaf, Iraq
| | | | - Zuhair Allebban
- Middle Euphrates Unit of Cancer Research, Kufa University College of Medicine, Al-Najaf, Iraq
| |
Collapse
|
32
|
Biswal NR, Mallick AK, Swain K, Sahoo JP. An Observational Study of Multi-Faceted Demyelinating Disorders. Cureus 2023; 15:e43775. [PMID: 37731435 PMCID: PMC10507210 DOI: 10.7759/cureus.43775] [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] [Accepted: 08/19/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Idiopathic inflammatory demyelinating diseases of the central nervous system (IIDCDs) are wide-ranging disorders due to their similarities and differences. In order to address these conditions, studying their characteristics is essential. The endpoints of our study were to assess the incidence, presenting features, MRI findings, and predictors of disease progression of prevalent demyelinating disorders. MATERIAL AND METHODS This prospective, observational study was conducted at Srirama Chandra Bhanja (SCB) Medical College and Hospital, India, from August 2018 to November 2021. Individuals of 18-65 years of age with common demyelinating disorders were assessed at baseline, six, 12, and 24 months. Univariate and multivariate analyses were performed for the assessment of predictors. We used R software (version 4.2.1; R Foundation for Statistical Computing, Vienna, Austria) for data analysis. RESULTS Two hundred twenty (79%) of 278 enrolled participants completed this study. The mean age of the study population was 52.3±11.4 years. One hundred thirty-eight (63%) of them were males. The most common IIDCD in our study was neuromyelitis optica spectrum disorder (NMOSD: 87, 39.5%), followed by multiple sclerosis (MS: 72, 32.7%), acute transverse myelitis (ATM: 35, 15.9%), and acute disseminated encephalomyelitis (ADEM: 26, 11.8%). The univariate analysis revealed that male gender, diabetes mellitus, and history of smoking or alcoholism were significant predictors of the disease progression. CONCLUSION The IIDCDs were polysymptomatic at the initial presentation. Male diabetics are more prone to progressive disorders. However, multivariate analysis did not provide statistically significant results.
Collapse
Affiliation(s)
- Nihar R Biswal
- Neurology, Srirama Chandra Bhanja (SCB) Medical College, Cuttack, IND
| | - Ashok K Mallick
- Neurology, Srirama Chandra Bhanja (SCB) Medical College, Cuttack, IND
| | - Kali Swain
- Neurology, Srirama Chandra Bhanja (SCB) Medical College, Cuttack, IND
| | | |
Collapse
|
33
|
Forero K, Buqaileh R, Sunderman C, AbouAlaiwi W. COVID-19 and Neurological Manifestations. Brain Sci 2023; 13:1137. [PMID: 37626493 PMCID: PMC10452375 DOI: 10.3390/brainsci13081137] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a newly emerged coronavirus, has sparked a global pandemic with its airborne transmission and ability to infect with asymptomatic patients. The pathophysiology is thought to relate to the binding of angiotensin converting enzyme 2 (ACE2) receptors in the body. These receptors are widely expressed in various body organs such as the lungs, the heart, the gastrointestinal tract (GIT), and the brain. This article reviews the current knowledge on the symptoms of coronavirus disease 2019 (COVID-19), highlighting the neurological symptoms that are associated with COVID-19, and discussing the possible mechanisms for SARS-CoV-2 virus infection in the body.
Collapse
Affiliation(s)
| | | | | | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (K.F.); (R.B.); (C.S.)
| |
Collapse
|
34
|
Giacco V, Flower G, Artamonova M, Hunter J, Padilla Requerey A, Hamilton NB. Transient receptor potential Ankyrin-1 (TRPA1) agonists suppress myelination and induce demyelination in organotypic cortical slices. Glia 2023; 71:1402-1413. [PMID: 36762504 PMCID: PMC10953362 DOI: 10.1002/glia.24347] [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: 07/01/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 02/11/2023]
Abstract
Oligodendrocytes are highly specialized glial cells characterized by their production of multilayer myelin sheaths that wrap axons to speed up action potential propagation. It is due to their specific role in supporting axons that impairment of myelin structure and function leads to debilitating symptoms in a wide range of degenerative diseases, including Multiple Sclerosis and Leukodystrophies. It is known that myelin damage can be receptor-mediated and recently oligodendrocytes have been shown to express Ca2+ -permeable Transient Receptor Potential Ankyrin-1 (TRPA1) channels, whose activation can result in myelin damage in ischemia. Here, we show, using organotypic cortical slice cultures, that TRPA1 activation, by TRPA1 agonists JT010 and Carvacrol for varying lengths of time, induces myelin damage. Although TRPA1 activation does not appear to affect oligodendrocyte progenitor cell number or proliferation, it prevents myelin formation and after myelination causes internodal shrinking and significant myelin degradation. This does not occur when the TRPA1 antagonist, A967079, is also applied. Of note is that when TRPA1 agonists are applied for either 24 h, 3 days or 7 days, axon integrity appears to be preserved while mature myelinated oligodendrocytes remain but with significantly shortened internodes. These results provide further evidence that TRPA1 inhibition could be protective in demyelination diseases and a promising therapy to prevent demyelination and promote remyelination.
Collapse
Affiliation(s)
- Vincenzo Giacco
- Wolfson Centre for Age‐Related DiseasesInstitute of Psychiatry, Psychology and Neuroscience, Guy's Campus, King's College LondonLondonUK
| | - Grace Flower
- Wolfson Centre for Age‐Related DiseasesInstitute of Psychiatry, Psychology and Neuroscience, Guy's Campus, King's College LondonLondonUK
| | - Maria Artamonova
- Wolfson Centre for Age‐Related DiseasesInstitute of Psychiatry, Psychology and Neuroscience, Guy's Campus, King's College LondonLondonUK
| | - Jake Hunter
- Wolfson Centre for Age‐Related DiseasesInstitute of Psychiatry, Psychology and Neuroscience, Guy's Campus, King's College LondonLondonUK
| | - Aitana Padilla Requerey
- Wolfson Centre for Age‐Related DiseasesInstitute of Psychiatry, Psychology and Neuroscience, Guy's Campus, King's College LondonLondonUK
| | - Nicola B. Hamilton
- Wolfson Centre for Age‐Related DiseasesInstitute of Psychiatry, Psychology and Neuroscience, Guy's Campus, King's College LondonLondonUK
| |
Collapse
|
35
|
Pratap Shankar KM, Pratibha PN, Saritha V. Ayurvedic management of neurological deficits post COVID-19 vaccination - A report of two cases. J Ayurveda Integr Med 2023; 14:100737. [PMID: 37343418 PMCID: PMC10247886 DOI: 10.1016/j.jaim.2023.100737] [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: 07/09/2022] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
The world witnessed much research fund allocation on the COVID-19 outbreak's epidemiology, pathology, impact on lifestyles, social behaviours and treatment possibilities. The highly contagious nature of the disease compelled scientific communities and related organisations to hasten vaccine development and supplies. Well-timed international collaborations resulted in quicker development of varied forms of vaccines against COVID-19. Prospective observational studies and systematic reviews on vaccine trials reported their safety and efficacies. Nevertheless, post-marketing surveillance is quintessential to ascertain such safety and efficacy claims. There have been scattered reports lately of several adverse temporal events, such as haematological, immunological and neurological untoward occurrences following COVID-19 inoculation. There is a growing piece of evidence of the impact of COVID vaccination on patients with neurological-neuroimmunological disorders. Here two unrelated cases of neurological deficits post-COVID vaccination are reported. One was an incidence of Acute Disseminated Encephalomyelitis, while the other was an acute exacerbation of Multiple Sclerosis following vaccination. Ayurvedic treatments were effective in either of these conditions. Case series and case reports shall judiciously add information to vaccine safety data and acknowledge the necessity of clinician approval, based on detailed individualised assessments before mass vaccination.
Collapse
Affiliation(s)
- K M Pratap Shankar
- National Ayurveda Research Institute for Panchakarma, Cheruthuruthy, Thrissur, Kerala, India.
| | - P Nair Pratibha
- Department Of Kayachikitsa, VPSV Ayurveda College, Kottakkal, Kerala, India
| | - V Saritha
- Department of Radiology, Government Medical College, Palakkad, Kerala, India
| |
Collapse
|
36
|
Phung NV, Rong F, Xia WY, Fan Y, Li XY, Wang SA, Li FL. Nervonic acid and its sphingolipids: Biological functions and potential food applications. Crit Rev Food Sci Nutr 2023; 64:8766-8785. [PMID: 37114919 DOI: 10.1080/10408398.2023.2203753] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Nervonic acid, a 24-carbon fatty acid with only one double bond at the 9th carbon (C24:1n-9), is abundant in the human brain, liver, and kidney. It not only functions in free form but also serves as a critical component of sphingolipids which participate in many biological processes such as cell membrane formation, apoptosis, and neurotransmission. Recent studies show that nervonic acid supplementation is not only beneficial to human health but also can improve the many medical conditions such as neurological diseases, cancers, diabetes, obesity, and their complications. Nervonic acid and its sphingomyelins serve as a special material for myelination in infants and remyelination patients with multiple sclerosis. Besides, the administration of nervonic acid is reported to reduce motor disorder in mice with Parkinson's disease and limit weight gain. Perturbations of nervonic acid and its sphingolipids might lead to the pathogenesis of many diseases and understanding these mechanisms is critical for investigating potential therapeutic approaches for such diseases. However, available studies about this aspect are limited. In this review, relevant findings about functional mechanisms of nervonic acid have been comprehensively and systematically described, focusing on four interconnected functions: cellular structure, signaling, anti-inflammation, lipid mobilization, and their related diseases.
Collapse
Affiliation(s)
- Nghi Van Phung
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Fei Rong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Wan Yue Xia
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Yong Fan
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Xian Yu Li
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Shi An Wang
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Shandong Energy Institute, Qingdao, China
| | - Fu Li Li
- Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
- Shandong Energy Institute, Qingdao, China
| |
Collapse
|
37
|
ELBini I, Neili NE. Potassium channels at the crossroads of neuroinflammation and myelination in experimental models of multiple sclerosis. Biochem Biophys Res Commun 2023; 653:140-146. [PMID: 36870238 DOI: 10.1016/j.bbrc.2023.02.066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS), characterized by the presence of localized demyelinating lesions accompanied by an inflammatory reaction, evidently leading to neurodegeneration. A number of ion channels have been implicated in the progression of MS, most notably in cell types involved in the immune response. In the present study, we investigated the implication of two ion channel isoforms, Kv1.1 and Kv1.3, in experimental models of neuroinflammation and demyelination. Immunohistochemical staining of brain sections from the mouse cuprizone model displayed high levels Kv1.3 expression. In an astroglial cellular model of inflammation, stimulation with LPS resulted also in a higher expression of Kv1.1 and Kv1.3, while the introduction of 4-Aminopyridine (4-AP) exacerbated the release of pro-inflammatory chemokine CXCL10. In the oligodendroglial cellular model of demyelination, the alteration in expression levels of Kv1.1 and Kv1.3 may be correlated with that of MBP levels. Indirect co-culture was attempted to further understand the communication between astrocytes and oligodendrocytes, The addition of reactive astrocytes' secretome significantly inhibited the production of MBP, this inhibition was accompanied by an alteration in the expression of Kv1.1 and Kv1.3. The addition of 4-AP in this case did not alleviate the decrease in MBP production. In conclusion, the use of 4-AP generated controversial results, suggesting 4-AP may be used in the early stages of the disease or in the remission phases to stimulate myelination, yet in induced toxic inflammatory environment, 4-AP exacerbated this effect.
Collapse
Affiliation(s)
- Ines ELBini
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, 1002, Tunisia.
| | - Nour-Elhouda Neili
- Laboratoire des Biomolécules, Venins et Applications Théranostiques (LR20IPT01), Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, 1002, Tunisia.
| |
Collapse
|
38
|
Berk-Rauch HE, Choudhury A, Richards AT, Singh PK, Chen ZL, Norris EH, Strickland S, Ahn HJ. Striatal fibrinogen extravasation and vascular degeneration correlate with motor dysfunction in an aging mouse model of Alzheimer’s disease. Front Aging Neurosci 2023; 15:1064178. [PMID: 36967821 PMCID: PMC10034037 DOI: 10.3389/fnagi.2023.1064178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction: Alzheimer’s Disease (AD) patients exhibit signs of motor dysfunction, including gait, locomotion, and balance deficits. Changes in motor function often precede other symptoms of AD as well as correlate with increased severity and mortality. Despite the frequent occurrence of motor dysfunction in AD patients, little is known about the mechanisms by which this behavior is altered.Methods and Results: In the present study, we investigated the relationship between cerebrovascular impairment and motor dysfunction in a mouse model of AD (Tg6799). We found an age-dependent increase of extravasated fibrinogen deposits in the cortex and striatum of AD mice. Interestingly, there was significantly decreased cerebrovascular density in the striatum of the 15-month-old as compared to 7-month-old AD mice. We also found significant demyelination and axonal damage in the striatum of aged AD mice. We analyzed striatum-related motor function and anxiety levels of AD mice at both ages and found that aged AD mice exhibited significant impairment of motor function but not in the younger AD mice.Discussion: Our finding suggests an enticing correlation between extravasated fibrinogen, cerebrovascular damage of the striatum, and motor dysfunction in an AD mouse model, suggesting a possible mechanism underlying motor dysfunction in AD.
Collapse
Affiliation(s)
- Hanna E. Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Arnab Choudhury
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Allison T. Richards
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Zu-Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Hyung Jin Ahn,
| |
Collapse
|
39
|
Nakano S, Uyeda A, Matsunaga YT, Muramatsu R. Phenotypic and transcriptional characterization of oligodendrocyte precursor cells in a 3D culture. Biomater Sci 2023; 11:2860-2869. [PMID: 36861675 DOI: 10.1039/d2bm01897g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Remyelination of the central nervous system (CNS) is a regenerative response that depends on the development of oligodendrocyte precursor cells (OPCs), which are generated from neural stem cells in developmental stages and exist as tissue stem cells in the adult CNS. Three-dimensional (3D) culture systems that recapitulate the complexity of the in vivo microenvironment are important for understanding the behavior of OPCs in remyelination and for exploring effective therapeutic approaches. In general, functional analysis of OPCs has mainly used two-dimensional (2D) culture systems; however, the differences between the properties of OPCs cultured in 2D and 3D have not been fully elucidated despite cellular functions being affected by the scaffold. In this study, we analyzed the phenotypic and transcriptomic differences in OPCs from 2D and collagen gel-based 3D cultures. In the 3D culture, the OPCs exhibited less than half ratio of proliferation and almost half ratio of differentiation to mature oligodendrocytes, compared to the 2D culture in the same culturing period. RNA-seq data showed robust changes in the expression level of genes associated with oligodendrocyte differentiation, and there were more up-regulated genes than down-regulated genes in 3D cultures compared to 2D cultures. In addition, the OPCs cultured in collagen gel scaffolds at lower collagen fiber densities showed higher proliferation activity compared with those cultured in collagen gel with higher collagen fiber densities. Our findings have identified the effect of culture dimension as well as the complexity of the scaffold on OPC responses at the cellular and molecular levels.
Collapse
Affiliation(s)
- Shizuka Nakano
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and, Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan. .,Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan. .,Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akiko Uyeda
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and, Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan.
| | - Yukiko T Matsunaga
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan. .,Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and, Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8502, Japan.
| |
Collapse
|
40
|
Baadsvik EL, Weiger M, Froidevaux R, Faigle W, Ineichen BV, Pruessmann KP. Mapping the myelin bilayer with short-T 2 MRI: Methods validation and reference data for healthy human brain. Magn Reson Med 2023; 89:665-677. [PMID: 36253953 PMCID: PMC10091754 DOI: 10.1002/mrm.29481] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/24/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE To explore the properties of short-T2 signals in human brain, investigate the impact of various experimental procedures on these properties and evaluate the performance of three-component analysis. METHODS Eight samples of non-pathological human brain tissue were subjected to different combinations of experimental procedures including D2 O exchange and frozen storage. Short-T2 imaging techniques were employed to acquire multi-TE (33-2067 μs) data, to which a three-component complex model was fitted in two steps to recover the properties of the underlying signal components and produce amplitude maps of each component. For validation of the component amplitude maps, the samples underwent immunohistochemical myelin staining. RESULTS The signal component representing the myelin bilayer exhibited super-exponential decay with T2,min of 5.48 μs and a chemical shift of 1.07 ppm, and its amplitude could be successfully mapped in both white and gray matter in all samples. These myelin maps corresponded well to myelin-stained tissue sections. Gray matter signals exhibited somewhat different components than white matter signals, but both tissue types were well represented by the signal model. Frozen tissue storage did not alter the signal components but influenced component amplitudes. D2 O exchange was necessary to characterize the non-aqueous signal components, but component amplitude mapping could be reliably performed also in the presence of H2 O signals. CONCLUSIONS The myelin mapping approach explored here produced reasonable and stable results for all samples. The extensive tissue and methodological investigations performed in this work form a basis for signal interpretation in future studies both ex vivo and in vivo.
Collapse
Affiliation(s)
- Emily Louise Baadsvik
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Markus Weiger
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Romain Froidevaux
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Wolfgang Faigle
- Neuroimmunology and MS Research Section, Neurology Clinic, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Benjamin Victor Ineichen
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Klaas Paul Pruessmann
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Balcaen T, Piens C, Mwema A, Chourrout M, Vandebroek L, Des Rieux A, Chauveau F, De Borggraeve WM, Hoffmann D, Kerckhofs G. Revealing the three-dimensional murine brain microstructure by contrast-enhanced computed tomography. Front Neurosci 2023; 17:1141615. [PMID: 37034159 PMCID: PMC10076597 DOI: 10.3389/fnins.2023.1141615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/08/2023] [Indexed: 04/11/2023] Open
Abstract
To improve our understanding of the brain microstructure, high-resolution 3D imaging is used to complement classical 2D histological assessment techniques. X-ray computed tomography allows high-resolution 3D imaging, but requires methods for enhancing contrast of soft tissues. Applying contrast-enhancing staining agents (CESAs) ameliorates the X-ray attenuating properties of soft tissue constituents and is referred to as contrast-enhanced computed tomography (CECT). Despite the large number of chemical compounds that have successfully been applied as CESAs for imaging brain, they are often toxic for the researcher, destructive for the tissue and without proper characterization of affinity mechanisms. We evaluated two sets of chemically related CESAs (organic, iodinated: Hexabrix and CA4+ and inorganic polyoxometalates: 1:2 hafnium-substituted Wells-Dawson phosphotungstate and Preyssler anion), for CECT imaging of healthy murine hemispheres. We then selected the CESA (Hexabrix) that provided the highest contrast between gray and white matter and applied it to a cuprizone-induced demyelination model. Differences in the penetration rate, effect on tissue integrity and affinity for tissue constituents have been observed for the evaluated CESAs. Cuprizone-induced demyelination could be visualized and quantified after Hexabrix staining. Four new non-toxic and non-destructive CESAs to the field of brain CECT imaging were introduced. The added value of CECT was shown by successfully applying it to a cuprizone-induced demyelination model. This research will prove to be crucial for further development of CESAs for ex vivo brain CECT and 3D histopathology.
Collapse
Affiliation(s)
- Tim Balcaen
- MolDesignS, Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Leuven, Belgium
- ContrasT Team, Institute of Mechanics, Materials and Civil Engineering, Mechatronic, Electrical Energy and Dynamic Systems, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Catherine Piens
- ContrasT Team, Institute of Mechanics, Materials and Civil Engineering, Mechatronic, Electrical Energy and Dynamic Systems, UCLouvain, Louvain-la-Neuve, Belgium
| | - Ariane Mwema
- Advanced Drug Delivery and Biomaterials, UCLouvain, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, UCLouvain, Brussels, Belgium
| | - Matthieu Chourrout
- Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR 5292, Bron, France
| | - Laurens Vandebroek
- Laboratory of Biomolecular Modelling and Design (LBMD), Biochemistry, Molecular and Structural Biology, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Anne Des Rieux
- Advanced Drug Delivery and Biomaterials, UCLouvain, Brussels, Belgium
| | - Fabien Chauveau
- Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre de Recherche en Neurosciences de Lyon U1028 UMR 5292, Bron, France
| | - Wim M. De Borggraeve
- MolDesignS, Sustainable Chemistry for Metals and Molecules, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Delia Hoffmann
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- ContrasT Team, Institute of Mechanics, Materials and Civil Engineering, Mechatronic, Electrical Energy and Dynamic Systems, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department Materials Engineering, KU Leuven, Leuven, Belgium
- *Correspondence: Greet Kerckhofs,
| |
Collapse
|
42
|
CNS Delivery of Nucleic Acid Therapeutics: Beyond the Blood-Brain Barrier and Towards Specific Cellular Targeting. Pharm Res 2023; 40:77-105. [PMID: 36380168 DOI: 10.1007/s11095-022-03433-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Nucleic acid-based therapeutic molecules including small interfering RNA (siRNA), microRNA(miRNA), antisense oligonucleotides (ASOs), messenger RNA (mRNA), and DNA-based gene therapy have tremendous potential for treating diseases in the central nervous system (CNS). However, achieving clinically meaningful delivery to the brain and particularly to target cells and sub-cellular compartments is typically very challenging. Mediating cell-specific delivery in the CNS would be a crucial advance that mitigates off-target effects and toxicities. In this review, we describe these challenges and provide contemporary evidence of advances in cellular and sub-cellular delivery using a variety of delivery mechanisms and alternative routes of administration, including the nose-to-brain approach. Strategies to achieve subcellular localization, endosomal escape, cytosolic bioavailability, and nuclear transfer are also discussed. Ultimately, there are still many challenges to translating these experimental strategies into effective and clinically viable approaches for treating patients.
Collapse
|
43
|
Nguyen AT, Kouri N, Labuzan SA, Przybelski SA, Lesnick TG, Raghavan S, Reid RI, Reichard RR, Knopman DS, Petersen RC, Jack CR, Mielke MM, Dickson DW, Graff-Radford J, Murray ME, Vemuri P. Neuropathologic scales of cerebrovascular disease associated with diffusion changes on MRI. Acta Neuropathol 2022; 144:1117-1125. [PMID: 35841412 PMCID: PMC9637622 DOI: 10.1007/s00401-022-02465-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 01/26/2023]
Abstract
Summarizing the multiplicity and heterogeneity of cerebrovascular disease (CVD) features into a single measure has been difficult in both neuropathology and imaging studies. The objective of this work was to evaluate the association between neuroimaging surrogates of CVD and two available neuropathologic CVD scales in those with both antemortem imaging CVD measures and postmortem CVD evaluation. Individuals in the Mayo Clinic Study of Aging with MRI scans within 5 years of death (N = 51) were included. Antemortem CVD measures were computed from diffusion MRI (dMRI), FLAIR, and T2* GRE imaging modalities and compared with postmortem neuropathologic findings using Kalaria and Strozyk Scales. Of all the neuroimaging measures, both regional and global dMRI measures were associated with Kalaria and Strozyk Scales (p < 0.05) and modestly correlated with global cognitive performance. The major conclusions from this study were: (i) microstructural white matter injury measurements using dMRI may be meaningful surrogates of neuropathologic CVD scales, because they aid in capturing diffuse (and early) changes to white matter and secondary neurodegeneration due to lesions; (ii) vacuolation in the corpus callosum may be associated with white matter changes measured on antemortem dMRI imaging; (iii) Alzheimer's disease neuropathologic change did not associate with neuropathologic CVD scales; and (iv) future work should be focused on developing better quantitative measures utilizing dMRI to optimally assess CVD-related neuropathologic changes.
Collapse
Affiliation(s)
- Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Sydney A Labuzan
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Sheelakumari Raghavan
- Department of Radiology, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA
| | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA
| | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic and Foundation, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
44
|
Baratta AM, Mangieri RA, Aziz HC, Lopez MF, Farris SP, Homanics GE. Effect of chronic intermittent ethanol vapor exposure on RNA content of brain-derived extracellular vesicles. Alcohol 2022; 105:9-24. [PMID: 36055466 PMCID: PMC10173183 DOI: 10.1016/j.alcohol.2022.08.006] [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: 03/17/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 01/26/2023]
Abstract
Extracellular vesicles (EVs) are important players in normal biological function and disease pathogenesis. Of the many biomolecules packaged into EVs, coding and noncoding RNA transcripts are of particular interest for their ability to significantly alter cellular and molecular processes. Here we investigate how chronic ethanol exposure impacts EV RNA cargo and the functional outcomes of these changes. Following chronic intermittent ethanol (CIE) vapor exposure, EVs were isolated from male and female C57BL/6J mouse brain. Total RNA from EVs was analyzed by lncRNA/mRNA microarray to survey changes in RNA cargo following vapor exposure. Differential expression analysis of microarray data revealed a number of lncRNA and mRNA types differentially expressed in CIE compared to control EVs. Weighted gene co-expression network analysis identified multiple male and female specific modules related to neuroinflammation, cell death, demyelination, and synapse organization. To functionally test these changes, whole-cell voltage-clamp recordings were used to assess synaptic transmission. Incubation of nucleus accumbens brain slices with EVs led to a reduction in spontaneous excitatory postsynaptic current amplitude, although no changes in synaptic transmission were observed between control and CIE EV administration. These results indicate that CIE vapor exposure significantly changes the RNA cargo of brain-derived EVs, which have the ability to impact neuronal function.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Regina A Mangieri
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Heather C Aziz
- College of Pharmacy, University of Texas at Austin, Texas, United States
| | - Marcelo F Lopez
- Department of Psychiatry and Behavioral Science, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Sean P Farris
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Gregg E Homanics
- Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States.
| |
Collapse
|
45
|
Putry BO, Khairunnisa N, Balga HM, Tjang YS, Thadeus MS, Santosa F, Pasiak TF. Can SARS-CoV-2 trigger new onset of autoimmune disease in adults? A case-based review. Heliyon 2022; 8:e11328. [PMCID: PMC9622433 DOI: 10.1016/j.heliyon.2022.e11328] [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: 03/22/2022] [Revised: 06/03/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Introduction Although it has been proposed that SARS-CoV-2 can cause autoimmunity by inducing a transient immunodeficiency of both innate and acquired immunity components in which the immune system fails to identify autoantigens adequately, the exact mechanism that causes this disease remains unknown. We aim to systematically review of existing case reports for evidence of new autoimmune diseases in adults caused by SARS-CoV-2 infection. Methods PRISMA-P 2020 method was used to search for literature in "PubMed" databases using the string "COVID-19 AND autoimmune disease AND complication". We used JBI Critical Appraisal Checklist to assess the articles' quality. Results The literature search yielded 666 articles. 58 articles met our eligibility criteria. Based on our critical appraisal, we placed 35 articles in the good category and 23 articles in the medium category. Data was synthesized by grouping similar data into a table, including: gender, age, COVID-19 severity, types of autoimmune diseases, autoimmune profile and relevant findings, when autoimmune diseases are diagnosed, complications, and outcome to draw conclusions. The new onset of autoimmune disease in adult triggered by SARS-CoV-2 included Guillain-Barré syndrome and Miller Fisher syndrome, systemic lupus erythematosus, immune thrombocytopenia, autoimmune haemolytic anemia, latent autoimmune diabetes in adults, myositis, acute demyelinating encephalomyelitis, autoimmune encephalitis, central nervous system vasculitis, and autoimmune thyroid diseases. Conclusion SARS-CoV-2 can trigger new onset of a variety of autoimmune diseases. Doctors who take care patients infected by COVID-19 must be aware of the complications of autoimmune diseases. Future cohort or cross-sectional studies on SARS-CoV-2-related autoimmune disease should be conducted.
Collapse
|
46
|
Werth R. A Scientific Approach to Conscious Experience, Introspection, and Unconscious Processing: Vision and Blindsight. Brain Sci 2022; 12:1305. [PMID: 36291239 PMCID: PMC9599441 DOI: 10.3390/brainsci12101305] [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: 08/06/2022] [Revised: 09/15/2022] [Accepted: 09/25/2022] [Indexed: 11/17/2022] Open
Abstract
Although subjective conscious experience and introspection have long been considered unscientific and banned from psychology, they are indispensable in scientific practice. These terms are used in scientific contexts today; however, their meaning remains vague, and earlier objections to the distinction between conscious experience and unconscious processing, remain valid. This also applies to the distinction between conscious visual perception and unconscious visual processing. Damage to the geniculo-striate pathway or the visual cortex results in a perimetrically blind visual hemifield contralateral to the damaged hemisphere. In some cases, cerebral blindness is not absolute. Patients may still be able to guess the presence, location, shape or direction of movement of a stimulus even though they report no conscious visual experience. This "unconscious" ability was termed "blindsight". The present paper demonstrates how the term conscious visual experience can be introduced in a logically precise and methodologically correct way and becomes amenable to scientific examination. The distinction between conscious experience and unconscious processing is demonstrated in the cases of conscious vision and blindsight. The literature on "blindsight" and its neurobiological basis is reviewed. It is shown that blindsight can be caused by residual functions of neural networks of the visual cortex that have survived cerebral damage, and may also be due to an extrastriate pathway via the midbrain to cortical areas such as areas V4 and MT/V5.
Collapse
Affiliation(s)
- Reinhard Werth
- Social Pediatrics and Adolescent Medicine, Ludwig-Maximilians-University of Munich, Haydnstr. 5, D-80336 München, Germany
| |
Collapse
|
47
|
Role of Demyelination in the Persistence of Neurological and Mental Impairments after COVID-19. Int J Mol Sci 2022; 23:ijms231911291. [PMID: 36232592 PMCID: PMC9569975 DOI: 10.3390/ijms231911291] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Long-term neurological and mental complications of COVID-19, the so-called post-COVID syndrome or long COVID, affect the quality of life. The most persistent manifestations of long COVID include fatigue, anosmia/hyposmia, insomnia, depression/anxiety, and memory/attention deficits. The physiological basis of neurological and psychiatric disorders is still poorly understood. This review summarizes the current knowledge of neurological sequelae in post-COVID patients and discusses brain demyelination as a possible mechanism of these complications with a focus on neuroimaging findings. Numerous reviews, experimental and theoretical studies consider brain demyelination as one of the mechanisms of the central neural system impairment. Several factors might cause demyelination, such as inflammation, direct effect of the virus on oligodendrocytes, and cerebrovascular disorders, inducing myelin damage. There is a contradiction between the solid fundamental basis underlying demyelination as the mechanism of the neurological injuries and relatively little published clinical evidence related to demyelination in COVID-19 patients. The reason for this probably lies in the fact that most clinical studies used conventional MRI techniques, which can detect only large, clearly visible demyelinating lesions. A very limited number of studies use specific methods for myelin quantification detected changes in the white matter tracts 3 and 10 months after the acute phase of COVID-19. Future research applying quantitative MRI assessment of myelin in combination with neurological and psychological studies will help in understanding the mechanisms of post-COVID complications associated with demyelination.
Collapse
|
48
|
Valenza M, Facchinetti R, Steardo L, Scuderi C. Palmitoylethanolamide and White Matter Lesions: Evidence for Therapeutic Implications. Biomolecules 2022; 12:biom12091191. [PMID: 36139030 PMCID: PMC9496237 DOI: 10.3390/biom12091191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
Palmitoylethanolamide (PEA), the naturally occurring amide of ethanolamine and palmitic acid, is an endogenous lipid compound endowed with a plethora of pharmacological functions, including analgesic, neuroprotective, immune-modulating, and anti-inflammatory effects. Although the properties of PEA were first characterized nearly 65 years ago, the identity of the receptor mediating these actions has long remained elusive, causing a period of research stasis. In the last two decades, a renewal of interest in PEA occurred, and a series of interesting studies have demonstrated the pharmacological properties of PEA and clarified its mechanisms of action. Recent findings showed the ability of formulations containing PEA in promoting oligodendrocyte differentiation, which represents the first step for the proper formation of myelin. This evidence opens new and promising research opportunities. White matter defects have been detected in a vast and heterogeneous group of diseases, including age-related neurodegenerative disorders. Here, we summarize the history and pharmacology of PEA and discuss its therapeutic potential in restoring white matter defects.
Collapse
Affiliation(s)
- Marta Valenza
- Department of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome—P.le A. Moro, 5, 00185 Rome, Italy
| | - Roberta Facchinetti
- Department of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome—P.le A. Moro, 5, 00185 Rome, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome—P.le A. Moro, 5, 00185 Rome, Italy
- Università Giustino Fortunato, 82100 Benevento, Italy
- Correspondence: (L.S.); (C.S.)
| | - Caterina Scuderi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, SAPIENZA University of Rome—P.le A. Moro, 5, 00185 Rome, Italy
- Correspondence: (L.S.); (C.S.)
| |
Collapse
|
49
|
Shim JM, Cho SE, Kang SG, Kang CK. Quantitative myelin-related maps from R1 and T2* ratio images using a single ME-MP2RAGE sequence in 7T MRI. Front Neuroanat 2022; 16:950650. [PMID: 36093293 PMCID: PMC9454012 DOI: 10.3389/fnana.2022.950650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There still are limitations associated with quantifying myelin content using brain magnetic resonance imaging (MRI) despite several studies conducted on this subject. Therefore, this study aimed: (1) to propose a myelin-related mapping technique to obtain the quantitative R1/T2* (q-Ratio) that has the advantage of quick processing and less dependency on imaging parameters, (2) to validate this adapted q-Ratio method by comparing the quantitative myelin-related map with those acquired through an existing mapping method [T1-weighted/T2*-weighted (w-Ratio)], and (3) to determine the q-Ratio myelin-related values in the white and gray matter, and the relationship between the q-Ratio myelin-related value and cerebral volume size in regions of interest (ROIs) in a healthy population.Methods: The multi-echo magnetization-prepared 2 rapid gradient echoes (ME-MP2RAGE) sequence was used in a 7 Tesla (7T) MRI for the acquisition of data regarding myelin content in 10 healthy participants. A correlation analysis was performed between myelin-related values obtained through the q-Ratio and w-Ratio methods. Additionally, myelin distribution was analyzed and compared in the white and gray matter, and the correlation between cerebral volume size and q-Ratio myelin-related value was analyzed in ROIs in the brain.Results: The myelin-related maps acquired through the q-Ratio and w-Ratio methods were significantly correlated (p < 0.001), but the q-Ratio myelin-related map was much clearer. Additionally, the cerebral volume size in the gray matter was 399.40% larger than that in the white matter, but the q-Ratio myelin-related value in the gray matter was 80.83% lower than that of the white matter. Furthermore, volume size was positively correlated with q-Ratio myelin-related values in the white matter (r = 0.509, p = 0.006) but not in the gray matter (r = -0.133, p = 0.402).Conclusions: In this study, we validated using a q-Ratio myelin-related map that was acquired in one imaging sequence at 7T MRI. In addition, we found a significant correlation between ROI volume size and the q-Ratio myelin-related value in the white matter but not in the gray matter. It is expected that this technique could be applied to the study of various neuropsychiatric diseases related to demyelination in the future.
Collapse
Affiliation(s)
- Jeong-Min Shim
- Neuroscience Research Institute, Gachon University, Incheon, South Korea
| | - Seo-Eun Cho
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Seung-Gul Kang
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
- *Correspondence: Seung-Gul Kang Chang-Ki Kang
| | - Chang-Ki Kang
- Neuroscience Research Institute, Gachon University, Incheon, South Korea
- Department of Radiological Science, College of Health Science, Gachon University, Incheon, South Korea
- *Correspondence: Seung-Gul Kang Chang-Ki Kang
| |
Collapse
|
50
|
Adebiyi O, Adigun K, David-Odewumi P, Akindele U, Olayemi F. Gallic and ascorbic acids supplementation alleviate cognitive deficits and neuropathological damage exerted by cadmium chloride in Wistar rats. Sci Rep 2022; 12:14426. [PMID: 36002551 PMCID: PMC9402671 DOI: 10.1038/s41598-022-18432-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Cadmium is a highly neurotoxic heavy metal that interferes with DNA repair mechanisms via generation of reactive oxygen species. The potentials of polyphenols and antioxidants as effective protective agents following heavy metal-induced neurotoxicity are emerging. We therefore explored the neuroprotective potentials of gallic and ascorbic acids in CdCl2-induced neurotoxicity. Seventy-two Wistar rats were divided into six groups. Group A received distilled water, B: 3 mg/kg CdCl2, C: 3 mg/kg CdCl2 + 20 mg/kg gallic acid (GA), D: 3 mg/kg CdCl2 + 10 mg/kg ascorbic acid (AA), E: 20 mg/kg GA and F: 10 mg/kg AA orally for 21 days. Depression, anxiety, locomotion, learning and memory were assessed using a battery of tests. Neuronal structure and myelin expression were assessed with histological staining and immunofluorescence. The Morris Water Maze test revealed significant increase in escape latency in CdCl2 group relative to rats concurrently treated with GA or AA. Similarly, time spent in the target quadrant was reduced significantly in CdCl2 group relative to other groups. Concomitant administration of gallic acid led to significant reduction in the durations of immobility and freezing that were elevated in CdCl2 group during forced swim and open field tests respectively. Furthermore, GA and AA restored myelin integrity and neuronal loss observed in the CdCl2 group. We conclude that gallic and ascorbic acids enhance learning and memory, decrease anxiety and depressive-like behavior in CdCl2-induced neurotoxicity with accompanying myelin-protective ability.
Collapse
Affiliation(s)
- Olamide Adebiyi
- Department of Veterinary Physiology and Biochemistry, University of Ibadan, Ibadan, Nigeria.
| | - Kabirat Adigun
- Department of Veterinary Physiology and Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Praise David-Odewumi
- Department of Veterinary Physiology and Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Uthman Akindele
- Department of Veterinary Physiology and Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Funsho Olayemi
- Department of Veterinary Physiology and Biochemistry, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|