1
|
Chen Z, He J, Guo Y, Hao Y, Lv W, Chen Z, Wang J, Yang Y, Wang K, Liu Z, Ouyang Q, Su Z, Hu P, Xiao G. Adherent junctions: Physiology, role in hydrocephalus and potential therapeutic targets. IBRO Neurosci Rep 2025; 18:283-292. [PMID: 39995568 PMCID: PMC11849119 DOI: 10.1016/j.ibneur.2025.02.003] [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: 11/30/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
In all epithelial cells, the adherent junctions (AJs) with cadherin as the core play an important role in the maintenance of the connection and the formation of apical-basal polarity. The ependymal cells close to the ventricular system rely on AJs with N-cadherin at the core to maintain their normal morphology and function. Therefore, it has an important impact on the function and disease of the central nervous system. Hydrocephalus is a pathological phenomenon of excessive cerebrospinal fluid accumulating in the ventricular system accompanied by continuous ventricular dilatation, which can be divided into obstructive hydrocephalus and communicating hydrocephalus according to the pathogenesis. Obstructive hydrocephalus is often associated with excessive ependymal cells produced by differentiation of radial glial cells. The etiology of communicating hydrocephalus is mainly related to the dyskinesia of cerebrospinal fluid. In addition, the damage of the brain barrier can lead to brain edema and aggravate the symptoms. At present, the researches on the pathogenesis of hydrocephalus are mainly focused on the development of ependymal cells and cilia, while less attention has been paid to molecules such as AJs, which play an important role in maintaining the polarity of ependymal cells. This paper discusses the formation and function of AJs and their role in preventing hydrocephalus by preserving the polarity of ependymal cilia, regulating the number of ependymal cells, and upholding the brain barrier integrity to impede hydrocephalus exacerbation, which provides a new direction for the study of hydrocephalus.
Collapse
Affiliation(s)
- Zhiye Chen
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410008, PR China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jian He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yating Guo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yue Hao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wentao Lv
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zexin Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Junqiang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Kaiyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qian Ouyang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Department of Neurosurgery, Zhuzhou Hospital, Central South University Xiangya School of Medicine, Zhuzhou, Hunan 412000, PR China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB21 2QQ, UK
| | - Pingsheng Hu
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410008, PR China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
2
|
Srivastava V, O'Reilly C. Characteristics of Cerebrospinal Fluid in Autism Spectrum Disorder - A Systematic Review. Neurosci Biobehav Rev 2025:106202. [PMID: 40354953 DOI: 10.1016/j.neubiorev.2025.106202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Autism Spectrum Disorder (ASD) is a range of neurodevelopmental conditions characterized by impaired social interaction, learning, and restricted or repetitive behaviors. The underlying causes of ASD are still debated, but researchers have found many physiological traits like gut problems and impaired immune system to help understand the etiology of ASD. Cerebrospinal fluid (CSF) plays a critical role in maintaining the homeostasis of the neuronal environment and has, therefore, been analyzed in multiple conditions impacting the central nervous system. The study of CSF is crucial to understanding neurological disorders as its composition changes with the disorders, and these changes may indicate various disorder-related physiological mechanisms. For this systematic review, we searched PubMed, Scopus, and Web of Science for studies published between 1977 and 2025 and selected 49 studies after manual screening. We took stock of the evidence supporting the hypothesis that ASD alters the properties and composition of CSF. We systematically report on the different attributes of CSF in the ASD population that could be potential biomarkers and assist in understanding the origins and progression of ASD. We found that in CSF, immune markers, proteins, extra-axial CSF, folate, oxytocin, and vasopressin showed changes in ASD compared to the neurotypicals. We observed gaps in the literature due to variations in age and sample size and noted biases related to sex (i.e., samples are predominantly including male participants) and age (i.e., a handful of studies were conducted on adults). Our review highlights the need for more research on CSF in ASD to improve our understanding of this disorder and identify CSF biomarkers.
Collapse
Affiliation(s)
- Vandana Srivastava
- AI Institute, University of South Carolina, 5th floor, 1112 Greene St., Columbia, South Carolina 29201, USA; Department of Computer Science and Engineering, University of South Carolina, 550 Assembly Street, Columbia, South Carolina 29201, USA; Carolina Autism and Neurodevelopment Research Center, University of South Carolina, 1800 Gervais Street, Columbia, South Carolina 29201, USA.
| | - Christian O'Reilly
- AI Institute, University of South Carolina, 5th floor, 1112 Greene St., Columbia, South Carolina 29201, USA; Department of Computer Science and Engineering, University of South Carolina, 550 Assembly Street, Columbia, South Carolina 29201, USA; Carolina Autism and Neurodevelopment Research Center, University of South Carolina, 1800 Gervais Street, Columbia, South Carolina 29201, USA; Institute for Mind and Brain, University of South Carolina, 1800 Gervais Street, Columbia, South Carolina 29201, USA.
| |
Collapse
|
3
|
Conti B, Di Napoli C, Hafdaoui S, Nicotra V, Cesaretti C, Runza L, Accurti V, Boito S, Iascone M, Marchetti D, Silipigni R, Finelli P, Natacci F. CTNND1-Related Disorder: New Insight on Prenatal Phenotype. Am J Med Genet A 2025; 197:e63921. [PMID: 39487033 DOI: 10.1002/ajmg.a.63921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/08/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
CTNND1 is a gene located in 11q12.1, encoding for p120 catenin, a protein involved in maintaining adherent junctions, regulating the epithelial-mesenchymal transition, and transcriptional signaling of different cellular pathways. Pathogenic variants in CTNND1 are classically associated with isolated cleft palate and Blefaro-cheilo-dontic syndrome, an autosomal dominant condition characterized by abnormalities of the eyelid. Considering different signs and symptoms associated first with Blefaro-cheilo-dontic syndrome and later specifically with CTNND1, Ahlaratani and colleagues proposed a wider developmental role for CTNND1 than previously described, associating a broader phenotypic spectrum. This report describes a prenatal case in which a CTNND1 pathogenic variant and reverse phenotyping allowed a diagnosis of Blefaro-cheilo-dontic syndrome associated with characteristics never related to Blefaro-cheilo-dontic syndrome or CTNND1, such as hydrocephalus. This report is the first detailed fetal case of Blefaro-cheilo-dontic syndrome, and the new feature reported is consistent with CTNND1 developmental role and may add new insights into the phenotype spectrum that is being defined.
Collapse
Affiliation(s)
- B Conti
- Biomedical and Clinical Science Department, University of Milan, Milan, Italy
| | - C Di Napoli
- Medical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - S Hafdaoui
- Biomedical and Clinical Science Department, University of Milan, Milan, Italy
| | - V Nicotra
- Medical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - C Cesaretti
- Medical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - L Runza
- Division of Pathology, Fondazione IRCCS ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - V Accurti
- Fetal Medicine and Surgery Service, Fondazione IRCCS ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - S Boito
- Fetal Medicine and Surgery Service, Fondazione IRCCS ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - M Iascone
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - D Marchetti
- Laboratory of Medical Genetics, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - R Silipigni
- Laboratory of Medical Genetics, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - P Finelli
- Laboratory of Medical Genetics, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - F Natacci
- Medical Genetics Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
4
|
Ma L, Liu S, Yao Q, Wang X, Guan J, Jian F. Ependymal cilia decline and AQP4 upregulation in young adult rats with syringomyelia. Fluids Barriers CNS 2025; 22:21. [PMID: 39994786 PMCID: PMC11849276 DOI: 10.1186/s12987-025-00631-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Syringomyelia was a rare condition characterized by the formation of fluid-filled cysts (syrinx) within the spinal cord, resulting in sensory and motor dysfunction. Currently, there was no satisfactory treatment for syringomyelia. Ependymal cells were integral to water transport and may represent a promising therapeutic target. METHODS Induction of syringomyelia occurred in 8-week old female rats followed by histological analyses at 3-, 7-, 14-, 30-, 60-, 180-, and 365-days later. Scanning electron microscope (SEM) and transmission electron microscope (TEM) were performed to visualize cilia on rat central canal membrane cells at 30-day post-induction. Syringomyelia was induced via compression at T12-T13 using a sterile cotton ball. Each rat underwent MRI scanning one day before induction and one day prior to sacrifice. In vivo magnetic resonance imaging (MRI) was utilized to measure syrinx enlargement in eight-week-old syringomyelia rats. Histological Analysis and immunofluorescence staining were performed for changes of cilia, neurons, expression of AQP4 and infiltration of immune cells into spinal tissue. RESULTS In the current study, the cell junctions between ependymal cells of syringomyelia rats were absent, and the cilia on ependymal cells were reduced significantly on day 30 post syringomyelia. The number of ependymal cells kept increasing lasting for 1-2 months and begin to decrease. Edema and vacuolation in the spinal cord tissue are significant in syringomyelia rats. Furthermore, AQP4 expression was elevated in astrocytes of syringomyelia rats, and IBA1+ immune cells infiltrated spinal tissue. Furthermore, neuronal necrosis began in the acute stage of syringomyelia, and reached its peak one month later. Pathological changes in axonal rupture at anterior commissure (connection of the left and right white matter) could be observed in syringomyelia spinal tissue. CONCLUSIONS These findings underscored the significance of cilia on ependymal cells and the evolving microenvironment post-syringomyelia, providing valuable insights for clinical treatment strategies for this condition.
Collapse
Affiliation(s)
- Longbing Ma
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Sumei Liu
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- Cell Therapy Center, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qingyu Yao
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Xinyu Wang
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Jian Guan
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Fengzeng Jian
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China.
- Lab of Spinal Cord Injury and Functional Reconstruction, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
- National Center for Neurological Disorders, Beijing, China.
| |
Collapse
|
5
|
Pang X, Gu L, Han QY, Xing JQ, Zhao M, Huang SY, Yi JX, Pan J, Hong H, Xue W, Zhou XQ, Su ZH, Zhang XR, Sun LM, Jiang SZ, Luo D, Chen L, Wang ZJ, Yu Y, Xia T, Zhang XM, Li AL, Zhou T, Cai H, Li T. RGS22 maintains the physiological function of ependymal cells to prevent hydrocephalus. SCIENCE CHINA. LIFE SCIENCES 2025; 68:441-453. [PMID: 39400871 DOI: 10.1007/s11427-024-2720-8] [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: 03/26/2024] [Accepted: 08/30/2024] [Indexed: 10/15/2024]
Abstract
Ependymal cells line the wall of cerebral ventricles and ensure the unidirectional cerebrospinal fluid (CSF) flow by beating their motile cilia coordinately. The ependymal denudation or ciliary dysfunction causes hydrocephalus. Here, we report that the deficiency of regulator of G-protein signaling 22 (RGS22) results in severe congenital hydrocephalus in both mice and rats. Interestingly, RGS22 is specifically expressed in ependymal cells within the brain. Using conditional knock-out mice, we further demonstrate that the deletion of Rgs22 exclusively in nervous system is sufficient to induce hydrocephalus. Mechanistically, we show that Rgs22 deficiency leads to the ependymal denudation and impaired ciliogenesis. This phenomenon can be attributed to the excessive activation of lysophosphatidic acid receptor (LPAR) signaling under Rgs22-/- condition, as the LPAR blockade effectively alleviates hydrocephalus in Rgs22-/- rats. Therefore, our findings unveil a previously unrecognized role of RGS22 in the central nervous system, and present RGS22 as a potential diagnostic and therapeutic target for hydrocephalus.
Collapse
Affiliation(s)
- Xue Pang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Lin Gu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jia-Qing Xing
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Ming Zhao
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Shao-Yi Huang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jun-Xi Yi
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jie Pan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Hao Hong
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Wen Xue
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xue-Qing Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Zhi-Hui Su
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xin-Ran Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Li-Ming Sun
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Shao-Zhen Jiang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dan Luo
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ling Chen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zheng-Jie Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Yu Yu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Tian Xia
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Hong Cai
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
| | - Tao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
6
|
Eisenhuth F, Agbonze JE, Groh AMR, Klostranec JM, Rudko DA, Stratton JA, Shapiro AJ. Age-related cerebral ventriculomegaly occurs in patients with primary ciliary dyskinesia. Fluids Barriers CNS 2025; 22:12. [PMID: 39891273 PMCID: PMC11783799 DOI: 10.1186/s12987-024-00614-9] [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: 10/05/2024] [Accepted: 12/20/2024] [Indexed: 02/03/2025] Open
Abstract
Primary ciliary dyskinesia (PCD) is a genetic disorder causing motile ciliary dysfunction primarily affecting the respiratory and reproductive systems. However, the impact of PCD on the central nervous system remains poorly understood. Rodent models of PCD exhibit marked hydrocephalus leading to early animal mortality, however, most humans with PCD do not develop hydrocephalus for unknown reasons. We hypothesized that patients with PCD exhibit sub-clinical ventriculomegaly related to ependymal motile ciliary dysfunction. We demonstrated highly specific expression levels of known PCD-related genes in human brain multiciliated ependymal cells (p < 0.0001). To assess ventricular size, computed tomography sinus images from patients with PCD (n = 33) and age/sex-matched controls (n = 64) were analysed. Patients with PCD displayed significantly larger ventricular areas (p < 0.0001) and Evans index (p < 0.01), indicating ventriculomegaly that was consistent across all genetic subgroups. Ventricular enlargement correlated positively with increasing age in patients with PCD compared to controls (p < 0.001). Additionally, chart review demonstrated a high prevalence (39%) of neuropsychiatric/neurological disorders in adult PCD patients that did not correlate with degree of ventriculomegaly. Our findings suggest that patients with PCD may have unrecognized, mild ventriculomegaly which correlates with ageing, potentially attributable to ependymal ciliary dysfunction. Further study is required to determine causality, and whether ventricular enlargement contributes to neuropsychiatric/neurological or other morbidity in PCD.
Collapse
Affiliation(s)
- Franziska Eisenhuth
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Joy E Agbonze
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Adam M R Groh
- Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Jesse M Klostranec
- Department of Neuroradiology, Montreal Neurological Institute and Hospital, Montreal, QC, H3A 2B4, Canada
- McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - David A Rudko
- Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 1A1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H3A 1A1, Canada.
- Montreal Neurological Institute, 3801 University Drive, Montreal, QC, H4A 3J1, Canada.
| | - Adam J Shapiro
- Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
- Montreal Children's Hospital, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
7
|
D'Gama PP, Jeong I, Nygård AM, Jamali A, Yaksi E, Jurisch-Yaksi N. Motile cilia modulate neuronal and astroglial activity in the zebrafish larval brain. Cell Rep 2025; 44:115195. [PMID: 39798091 DOI: 10.1016/j.celrep.2024.115195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 10/11/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
The brain uses a specialized system to transport cerebrospinal fluid (CSF), consisting of interconnected ventricles lined by motile ciliated ependymal cells. These cells act jointly with CSF secretion and cardiac pressure gradients to regulate CSF dynamics. To date, the link between cilia-mediated CSF flow and brain function is poorly understood. Using zebrafish larvae as a model system, we identify that loss of ciliary motility does not alter progenitor proliferation, brain morphology, or spontaneous neural activity despite leading to an enlarged telencephalic ventricle. We observe altered neuronal responses to photic stimulations in the optic tectum and hindbrain and brain asymmetry defects in the habenula. Finally, we investigate astroglia since they contact CSF and regulate neuronal activity. Our analyses reveal a reduction in astroglial calcium signals during both spontaneous and light-evoked activity. Our findings highlight a role of motile cilia in regulating brain physiology through the modulation of neural and astroglial networks.
Collapse
Affiliation(s)
- Percival P D'Gama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway
| | - Inyoung Jeong
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway
| | - Andreas Moe Nygård
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway
| | - Ahmed Jamali
- Kavli Institute for Systems Neuroscience and Centre for Algorithms in the Cortex, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Algorithms in the Cortex, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway; Koç University Research Center for Translational Medicine, Koç University School of Medicine, Davutpaşa Caddesi, No:4, Topkapı, Istanbul 34010, Turkey
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgssons Gate 1, 7491 Trondheim, Norway; Kavli Institute for Systems Neuroscience and Centre for Algorithms in the Cortex, Norwegian University of Science and Technology, Olav Kyrres Gate 9, 7030 Trondheim, Norway.
| |
Collapse
|
8
|
Rahmani D, Taheri RA, Moosazadeh Moghaddam M. Targeted delivery of curcumin and CM11 peptide against hepatocellular carcinoma cells based on binding affinity of PreS1-coated chitosan nanoparticles to SB3 protein. Amino Acids 2025; 57:12. [PMID: 39862295 PMCID: PMC11762422 DOI: 10.1007/s00726-024-03438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025]
Abstract
In recent years, the use of cationic peptides as alternative drugs with anticancer activity has received attention. In this study, the targeted release of curcumin (Cur) and CM11 peptide alone and together against hepatocellular carcinoma (HCC) was evaluated using chitosan nanoparticles (CS NPs) coated with Pres1 that target the SB3 antigen of HCC cells (PreS1-Cur-CM11-CS NPs). SB3 protein is the specific antigen of HCC and the PreS1 peptide is a part of the hepatitis B antigen, which can specifically bind to the SB3 protein. Chitosan was used to prepare NPs. To Cur and CM11 loading, drugs were added to the CS solution in appropriate concentrations. Pres1 was coupled to the surface of the NPs using EDC catalyst to target NPs against HepG2 cells. SEM and DLS analysis confirmed that the PreS1-Cur-CM11-CS NPs had a size of about 132 nm, the ideal size for penetrating the cell membrane. The loading of Cur and CM11 was equal to 87% and 65%, respectively, which had a sustained and better release in the acidic environment than in the physiological environment. The MTT assay showed that PreS1-Cur-CM11-CS NPs act in a targeted and specific manner with the highest toxicity on the HepG2 cells compared to the control by a decrease in viability of about 26% after 48 h based on cell apoptosis. The results showed that PreS1-Cur-CM11-CS NPs are capable of targeted and specific drug release against HepG2 cancer cells and have significant potential to fight this cancer.
Collapse
Affiliation(s)
- Danial Rahmani
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Wu W, Li Q. Mechanisms of hydrocephalus after intraventricular haemorrhage: a review. Childs Nerv Syst 2024; 41:49. [PMID: 39674974 DOI: 10.1007/s00381-024-06711-2] [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: 09/26/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Intraventricular haemorrhage (IVH) is bleeding within the ventricular system, which in adults is usually mainly secondary to cerebral haemorrhage and subarachnoid haemorrhage. Hydrocephalus is one of the most common complications of intraventricular haemorrhage, which is characterised by an increase in intracranial pressure due to an increased accumulation of cerebrospinal fluid within the ventricular system, and is closely related to the patient's prognosis. Surgical methods such as shunt surgery have been used to treat secondary hydrocephalus in recent years and have been effective in improving the survival and prognosis of patients with hydrocephalus. However, complications such as shunt blockage and intracranial infection are often faced after surgery. Moreover, little is known about the mechanism of hydrocephalus secondary to intraventricular haemorrhage. This review discusses the mechanisms regarding the occurrence of secondary hydrocephalus after intraventricular haemorrhage in adults in terms of blood clot obstruction, altered cerebrospinal fluid dynamics, inflammation, and blood composition.
Collapse
Affiliation(s)
- Wenchao Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 157, Health Care RoadHeilongjiang Province, Harbin City, Harbin, China
| | - Qingsong Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, No. 157, Health Care RoadHeilongjiang Province, Harbin City, Harbin, China.
| |
Collapse
|
10
|
Kaplani K, Lalioti ME, Vassalou S, Lokka G, Parlapani E, Kritikos G, Lygerou Z, Taraviras S. Ependymal cell lineage reprogramming as a potential therapeutic intervention for hydrocephalus. EMBO Mol Med 2024; 16:2725-2748. [PMID: 39468302 PMCID: PMC11555118 DOI: 10.1038/s44321-024-00156-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 09/22/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Hydrocephalus is a common neurological condition, characterized by the excessive accumulation of cerebrospinal fluid in the cerebral ventricles. Primary treatments for hydrocephalus mainly involve neurosurgical cerebrospinal fluid diversion, which hold high morbidity and failure rates, highlighting the necessity for the discovery of novel therapeutic approaches. Although the pathophysiology of hydrocephalus is highly multifactorial, impaired function of the brain ependymal cells plays a fundamental role in hydrocephalus. Here we show that GemC1 and McIdas, key regulators of multiciliated ependymal cell fate determination, induce direct cellular reprogramming towards ependyma. Our study reveals that ectopic expression of GemC1 and McIdas reprograms cortical astrocytes and programs mouse embryonic stem cells into ependyma. McIdas is sufficient to establish functional activity in the reprogrammed astrocytes. Furthermore, we show that McIdas' expression promotes ependymal cell regeneration in two different postnatal hydrocephalus mouse models: an intracranial hemorrhage and a genetic form of hydrocephalus and ameliorates the cytoarchitecture of the neurogenic niche. Our study provides evidence on the restoration of ependyma in animal models mimicking hydrocephalus that could be exploited towards future therapeutic interventions.
Collapse
Affiliation(s)
- Konstantina Kaplani
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Maria-Eleni Lalioti
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Styliani Vassalou
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Georgia Lokka
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Evangelia Parlapani
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Georgios Kritikos
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, Patras, Greece.
| |
Collapse
|
11
|
Xie S, Li F. Ependymal cells: roles in central nervous system infections and therapeutic application. J Neuroinflammation 2024; 21:255. [PMID: 39385253 PMCID: PMC11465851 DOI: 10.1186/s12974-024-03240-2] [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: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.
Collapse
Affiliation(s)
- Shiqi Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, 130 Dong An Road, Xuhui District, Shanghai, China.
- Tuberculosis Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
12
|
Herman J, Rittenhouse N, Mandino F, Majid M, Wang Y, Mezger A, Kump A, Kadian S, Lake EMR, Verardi PH, Conover JC. Ventricular-subventricular zone stem cell niche adaptations in a mouse model of post-infectious hydrocephalus. Front Neurosci 2024; 18:1429829. [PMID: 39145299 PMCID: PMC11322059 DOI: 10.3389/fnins.2024.1429829] [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: 05/08/2024] [Accepted: 07/02/2024] [Indexed: 08/16/2024] Open
Abstract
Congenital post-infectious hydrocephalus (PIH) is a condition characterized by enlargement of the ventricular system, consequently imposing a burden on the associated stem cell niche, the ventricular-subventricular zone (V-SVZ). To investigate how the V-SVZ adapts in PIH, we developed a mouse model of influenza virus-induced PIH based on direct intracerebroventricular injection of mouse-adapted influenza virus at two distinct time points: embryonic day 16 (E16), when stem cells line the ventricle, and postnatal day 4 (P4), when an ependymal monolayer covers the ventricle surface and stem cells retain only a thin ventricle-contacting process. Global hydrocephalus with associated regions of astrogliosis along the lateral ventricle was found in 82% of the mice infected at P4. Increased ependymogenesis was observed at gliotic borders and throughout areas exhibiting intact ependyma based on tracking of newly divided cells. Additionally, in areas of intact ependyma, stem cell numbers were reduced; however, we found no significant reduction in new neurons reaching the olfactory bulb following onset of ventriculomegaly. At P4, injection of only the non-infectious viral component neuraminidase resulted in limited, region-specific ventriculomegaly due to absence of cell-to-cell transmission. In contrast, at E16 intracerebroventricular injection of influenza virus resulted in death at birth due to hypoxia and multiorgan hemorrhage, suggesting an age-dependent advantage in neonates, while the viral component neuraminidase resulted in minimal, or no, ventriculomegaly. In summary, we tracked acute adaptations of the V-SVZ stem cell niche following onset of ventriculomegaly and describe developmental changes that help mitigate the severity of congenital PIH.
Collapse
Affiliation(s)
- Julianna Herman
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Nicole Rittenhouse
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Francesca Mandino
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States
| | - Mushirah Majid
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Yuxiang Wang
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT, United States
| | - Amelia Mezger
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Aidan Kump
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Sumeet Kadian
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| | - Evelyn M. R. Lake
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Wu Tsai Institute, Yale University, New Haven, CT, United States
| | - Paulo H. Verardi
- Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT, United States
| | - Joanne C. Conover
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
13
|
Xie S, Xie X, Tang J, Luo B, Chen J, Wen Q, Zhou J, Chen G. Cerebral furin deficiency causes hydrocephalus in mice. Genes Dis 2024; 11:101009. [PMID: 38292192 PMCID: PMC10825277 DOI: 10.1016/j.gendis.2023.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/20/2023] [Accepted: 04/29/2023] [Indexed: 02/01/2024] Open
Abstract
Furin is a pro-protein convertase that moves between the trans-Golgi network and cell surface in the secretory pathway. We have previously reported that cerebral overexpression of furin promotes cognitive functions in mice. Here, by generating the brain-specific furin conditional knockout (cKO) mice, we investigated the role of furin in brain development. We found that furin deficiency caused early death and growth retardation. Magnetic resonance imaging showed severe hydrocephalus. In the brain of furin cKO mice, impaired ciliogenesis and the derangement of microtubule structures appeared along with the down-regulated expression of RAB28, a ciliary vesicle protein. In line with the widespread neuronal loss, ependymal cell layers were damaged. Further proteomics analysis revealed that cell adhesion molecules including astrocyte-enriched ITGB8 and BCAR1 were altered in furin cKO mice; and astrocyte overgrowth was accompanied by the reduced expression of SOX9, indicating a disrupted differentiation into ependymal cells. Together, whereas alteration of RAB28 expression correlated with the role of vesicle trafficking in ciliogenesis, dysfunctional astrocytes might be involved in ependymal damage contributing to hydrocephalus in furin cKO mice. The structural and molecular alterations provided a clue for further studying the potential mechanisms of furin.
Collapse
Affiliation(s)
- Shiqi Xie
- Nursing College, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoyong Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Jing Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Biao Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Jian Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Qixin Wen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Jianrong Zhou
- Nursing College, Chongqing Medical University, Chongqing 400016, China
| | - Guojun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| |
Collapse
|
14
|
Jackson RJ, Keiser MS, Meltzer JC, Fykstra DP, Dierksmeier SE, Hajizadeh S, Kreuzer J, Morris R, Melloni A, Nakajima T, Tecedor L, Ranum PT, Carrell E, Chen Y, Nishtar MA, Holtzman DM, Haas W, Davidson BL, Hyman BT. APOE2 gene therapy reduces amyloid deposition and improves markers of neuroinflammation and neurodegeneration in a mouse model of Alzheimer disease. Mol Ther 2024; 32:1373-1386. [PMID: 38504517 PMCID: PMC11081918 DOI: 10.1016/j.ymthe.2024.03.024] [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: 07/17/2023] [Revised: 01/05/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do, they have a later age of onset, milder clinical course, and less severe neuropathological findings than people without this allele. The contrast is especially stark when compared with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, a more aggressive clinical course and more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Here, we demonstrate that brain exposure to APOE ε2 via a gene therapy approach, which bathes the entire cortical mantle in the gene product after transduction of the ependyma, reduces Aβ plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE ε4. This result suggests a promising protective effect of exogenous APOE ε2 and reveals a cell nonautonomous effect of the protein on microglial activation, which we show is similar to plaque-associated microglia in the brain of Alzheimer disease patients who inherit APOE ε2. These data increase the potential that an APOE ε2 therapeutic could be effective in Alzheimer disease, even in individuals born with the risky ε4 allele.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Megan S Keiser
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jonah C Meltzer
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Dustin P Fykstra
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Steven E Dierksmeier
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA; Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Soroush Hajizadeh
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Johannes Kreuzer
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Morris
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK
| | - Alexandra Melloni
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Tsuneo Nakajima
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Luis Tecedor
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul T Ranum
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ellie Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - YongHong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maryam A Nishtar
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Wilhelm Haas
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
15
|
Bork PAR, Hauglund NL, Mori Y, Møllgård K, Hjorth PG, Nedergaard M. Modeling of brain efflux: Constraints of brain surfaces. Proc Natl Acad Sci U S A 2024; 121:e2318444121. [PMID: 38598340 PMCID: PMC11032467 DOI: 10.1073/pnas.2318444121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/04/2024] [Indexed: 04/12/2024] Open
Abstract
Fluid efflux from the brain plays an important role in solute waste clearance. Current experimental approaches provide little spatial information, and data collection is limited due to short duration or low frequency of sampling. One approach shows tracer efflux to be independent of molecular size, indicating bulk flow, yet also decelerating like simple membrane diffusion. In an apparent contradiction to this report, other studies point to tracer efflux acceleration. We here develop a one-dimensional advection-diffusion model to gain insight into brain efflux principles. The model is characterized by nine physiological constants and three efflux parameters for which we quantify prior uncertainty. Using Bayes' rule and the two efflux studies, we validate the model and calculate data-informed parameter distributions. The apparent contradictions in the efflux studies are resolved by brain surface boundaries being bottlenecks for efflux. To critically test the model, a custom MRI efflux assay measuring solute dispersion in tissue and release to cerebrospinal fluid was employed. The model passed the test with tissue bulk flow velocities in the range 60 to 190 [Formula: see text]m/h. Dimensional analysis identified three principal determinants of efflux, highlighting brain surfaces as a restricting factor for metabolite solute clearance.
Collapse
Affiliation(s)
- Peter A. R. Bork
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200Denmark
| | - Natalie L. Hauglund
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200Denmark
| | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200Denmark
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200Denmark
| | - Poul G. Hjorth
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby2800Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen2200Denmark
| |
Collapse
|
16
|
Luckmann MR, Ferreira MAS, da Silva NM, Nazari EM. Developmental toxicity of pyriproxyfen induces changes in the ultrastructure of neural cells and in the process of skull ossification. Toxicol Sci 2024; 198:260-272. [PMID: 38268486 DOI: 10.1093/toxsci/kfae009] [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] [Indexed: 01/26/2024] Open
Abstract
Some studies relate the use of pyriproxyfen (PPF) in drinking water with damage to embryonic neurodevelopment, including a supposed association with cases of microcephaly. However, the effects on neural cells and skull ossification in embryos remain unclear. This study aims to investigate the effects of PPF on the structure and ultrastructure of brain cells and its influence on the skull ossification process during embryonic development. Chicken embryos, used as an experimental model, were exposed to concentrations of 0.01 and 10 mg/l PPF at E1. The findings demonstrated that PPF led to notable ultrastructural alterations such as reduced cilia and microvilli of ependymal cells and damage to mitochondria, endoplasmic reticulum, Golgi bodies, and cell membranes in neural cells. The frequency of changes and the degree of these cell damage between the forebrain and midbrain were similar. PPF induced a reduction in fox3 transcript levels, specific for differentiation of neurons, and a reduction in the NeuN protein content related to mature neurons and dendritic branches. PPF impacted the ossification process of the skull, as evidenced by the increase in the ossified area and the decrease in inter-bone spacing. In conclusion, this study highlights the ability of PPF to affect neurodevelopmental processes by inducing ultrastructural damage to neural cells, concomitant with a reduction in NeuN and fox3 expression. This detrimental impact coupled with deficiencies in skull ossification can prevent the proper growth and development of the brain.
Collapse
Affiliation(s)
- Maico Roberto Luckmann
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Méllanie Amanda Silva Ferreira
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Norma Machado da Silva
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Evelise Maria Nazari
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| |
Collapse
|
17
|
Rodriguez-Perez LM, Ojeda-Pérez B, López-de-San-Sebastián J, García-Bonilla M, González-García M, Fernández-Muñoz B, Sánchez-Pernaute R, García-Martín ML, Domínguez-Pinos D, Cárdenas-García C, Jiménez AJ, Paez-Gonzalez P. Design of a Stem Cell-Based Therapy for Ependymal Repair in Hydrocephalus Associated With Germinal Matrix Hemorrhages. Stroke 2024; 55:1062-1074. [PMID: 38436063 PMCID: PMC10962438 DOI: 10.1161/strokeaha.123.044677] [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: 05/25/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND In preterm birth germinal matrix hemorrhages (GMHs) and the consequent posthemorrhagic hydrocephalus (PHH), the neuroepithelium/ependyma development is disrupted. This work is aimed to explore the possibilities of ependymal repair in GMH/PHH using a combination of neural stem cells, ependymal progenitors (EpPs), and mesenchymal stem cells. METHODS GMH/PHH was induced in 4-day-old mice using collagenase, blood, or blood serum injections. PHH severity was characterized 2 weeks later using magnetic resonance, immunofluorescence, and protein expression quantification with mass spectrometry. Ependymal restoration and wall regeneration after stem cell treatments were tested in vivo and in an ex vivo experimental approach using ventricular walls from mice developing moderate and severe GMH/PHH. The effect of the GMH environment on EpP differentiation was tested in vitro. Two-tailed Student t or Wilcoxon-Mann-Whitney U test was used to find differences between the treated and nontreated groups. ANOVA and Kruskal-Wallis tests were used to compare >2 groups with post hoc Tukey and Dunn multiple comparison tests, respectively. RESULTS PHH severity was correlated with the extension of GMH and ependymal disruption (means, 88.22% severe versus 19.4% moderate). GMH/PHH hindered the survival rates of the transplanted neural stem cells/EpPs. New multiciliated ependymal cells could be generated from transplanted neural stem cells and more efficiently from EpPs (15% mean increase). Blood and TNFα (tumor necrosis factor alpha) negatively affected ciliogenesis in cells committed to ependyma differentiation (expressing Foxj1 [forkhead box J1] transcription factor). Pretreatment with mesenchymal stem cells improved the survival rates of EpPs and ependymal differentiation while reducing the edematous (means, 18% to 0.5% decrease in severe edema) and inflammatory conditions in the explants. The effectiveness of this therapeutical strategy was corroborated in vivo (means, 29% to 0% in severe edema). CONCLUSIONS In GMH/PHH, the ependyma can be restored and edema decreased from either neural stem cell or EpP transplantation in vitro and in vivo. Mesenchymal stem cell pretreatment improved the success of the ependymal restoration.
Collapse
Affiliation(s)
- Luis M Rodriguez-Perez
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, University of Malaga, Spain. (L.M.R.-P.)
| | - Betsaida Ojeda-Pérez
- Departamento de Biología Celular, Genética y Fisiología, University of Malaga, Spain. (B.O.-P., J.L.-d.-S.-S., M.G.-G.)
- Instituto de Investigación Biomédica de Málaga, Spain (B.O.-P., M.L.G.-M., D.D.-P., A.J.J., P.P.-G.)
| | - Javier López-de-San-Sebastián
- Departamento de Biología Celular, Genética y Fisiología, University of Malaga, Spain. (B.O.-P., J.L.-d.-S.-S., M.G.-G.)
| | - María García-Bonilla
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, MO (M.G.-B.)
| | - Marcos González-García
- Departamento de Biología Celular, Genética y Fisiología, University of Malaga, Spain. (B.O.-P., J.L.-d.-S.-S., M.G.-G.)
| | - Beatriz Fernández-Muñoz
- Unidad de Producción y Reprogramación Celular, Red Andaluza para el diseño y traslación de Terapias Avanzadas, Sevilla, Spain (B.F.-M.)
| | - Rosario Sánchez-Pernaute
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain (R.S.-P.)
- Instituto de Investigación Sanitaria Biobizkai, Barakaldo, Spain (R.S.-P.)
| | - María L García-Martín
- Instituto de Investigación Biomédica de Málaga, Spain (B.O.-P., M.L.G.-M., D.D.-P., A.J.J., P.P.-G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina, Spain (M.L.G.-M.)
| | - Dolores Domínguez-Pinos
- Departamento de Radiología y Medicina Física, Oftalmología y Otorrinolaringología, University of Malaga, Spain. (D.D.-P.)
- Instituto de Investigación Biomédica de Málaga, Spain (B.O.-P., M.L.G.-M., D.D.-P., A.J.J., P.P.-G.)
| | | | - Antonio J Jiménez
- Instituto de Investigación Biomédica de Málaga, Spain (B.O.-P., M.L.G.-M., D.D.-P., A.J.J., P.P.-G.)
| | - Patricia Paez-Gonzalez
- Instituto de Investigación Biomédica de Málaga, Spain (B.O.-P., M.L.G.-M., D.D.-P., A.J.J., P.P.-G.)
| |
Collapse
|
18
|
Preziosa P, Pagani E, Meani A, Storelli L, Margoni M, Yudin Y, Tedone N, Biondi D, Rubin M, Rocca MA, Filippi M. Chronic Active Lesions and Larger Choroid Plexus Explain Cognition and Fatigue in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200205. [PMID: 38350048 DOI: 10.1212/nxi.0000000000200205] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/18/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND AND OBJECTIVES Chronic inflammation may contribute to cognitive dysfunction and fatigue in patients with multiple sclerosis (MS). Paramagnetic rim lesions (PRLs) and choroid plexus (CP) enlargement have been proposed as markers of chronic inflammation in MS being associated with a more severe disease course. However, their relation with cognitive impairment and fatigue has not been fully explored yet. Here, we investigated the contribution of PRL number and volume and CP enlargement to cognitive impairment and fatigue in patients with MS. METHODS Brain 3T MRI, neurologic evaluation, and neuropsychological assessment, including the Brief Repeatable Battery of Neuropsychological Tests and Modified Fatigue Impact Scale, were obtained from 129 patients with MS and 73 age-matched and sex-matched healthy controls (HC). PRLs were identified on phase images of susceptibility-weighted imaging, whereas CP volume was quantified using a fully automatic method on brain three-dimensional T1-weighted and fluid-attenuated inversion recovery MRI sequences. Predictors of cognitive impairment and fatigue were identified using random forest. RESULTS Thirty-six (27.9%) patients with MS were cognitively impaired, and 31/113 (27.4%) patients had fatigue. Fifty-nine (45.7%) patients with MS had ≥1 PRLs (median = 0, interquartile range = 0;2). Compared with HC, patients with MS showed significantly higher T2-hyperintense white matter lesion (WM) volume; lower normalized brain, thalamic, hippocampal, caudate, cortical, and WM volumes; and higher normalized CP volume (p from <0.001 to 0.040). The predictors of cognitive impairment (relative importance) (out-of-bag area under the curve [OOB-AUC] = 0.707) were normalized brain volume (100%), normalized caudate volume (89.1%), normalized CP volume (80.3%), normalized cortical volume (70.3%), number (67.3%) and volume (66.7%) of PRLs, and T2-hyperintense WM lesion volume (64.0%). Normalized CP volume was the only predictor of the presence of fatigue (OOB-AUC = 0.563). DISCUSSION Chronic inflammation, with higher number and volume of PRLs and enlarged CP, may contribute to cognitive impairment in MS in addition to gray matter atrophy. The contribution of enlarged CP in explaining fatigue supports the relevance of immune-related processes in determining this manifestation independently of disease severity. PRLs and CP enlargement may contribute to the pathophysiology of cognitive impairment and fatigue in MS, and they may represent clinically relevant therapeutic targets to limit the impact of these clinical manifestations in MS.
Collapse
Affiliation(s)
- Paolo Preziosa
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Meani
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Storelli
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Margoni
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Yury Yudin
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicolò Tedone
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Diana Biondi
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Rubin
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- From the Neuroimaging Research Unit (P.P., E.P., A.M., L.S., M.M., Y.Y., N.T., D.B., M.R., M.A.R., M.F.), Division of Neuroscience; Neurology Unit (P.P., M.M., M.R., M.A.R., M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (P.P., M.R., M.A.R., M.F.); Neurorehabilitation Unit (M.M., M.F.); and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
19
|
Rodríguez-Pérez LM, López-de-San-Sebastián J, de Diego I, Smith A, Roales-Buján R, Jiménez AJ, Paez-Gonzalez P. A selective defect in the glial wedge as part of the neuroepithelium disruption in hydrocephalus development in the mouse hyh model is associated with complete corpus callosum dysgenesis. Front Cell Neurosci 2024; 18:1330412. [PMID: 38450283 PMCID: PMC10915275 DOI: 10.3389/fncel.2024.1330412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Dysgenesis of the corpus callosum is present in neurodevelopmental disorders and coexists with hydrocephalus in several human congenital syndromes. The mechanisms that underlie the etiology of congenital hydrocephalus and agenesis of the corpus callosum when they coappear during neurodevelopment persist unclear. In this work, the mechanistic relationship between both disorders is investigated in the hyh mouse model for congenital hydrocephalus, which also develops agenesis of the corpus callosum. In this model, hydrocephalus is generated by a defective program in the development of neuroepithelium during its differentiation into radial glial cells. Methods In this work, the populations implicated in the development of the corpus callosum (callosal neurons, pioneering axons, glial wedge cells, subcallosal sling and indusium griseum glial cells) were studied in wild-type and hyh mutant mice. Immunohistochemistry, mRNA in situ hybridization, axonal tracing experiments, and organotypic cultures from normal and hyh mouse embryos were used. Results Our results show that the defective program in the neuroepithelium/radial glial cell development in the hyh mutant mouse selectively affects the glial wedge cells. The glial wedge cells are necessary to guide the pioneering axons as they approach the corticoseptal boundary. Our results show that the pioneering callosal axons arising from neurons in the cingulate cortex can extend projections to the interhemispheric midline in normal and hyh mice. However, pioneering axons in the hyh mutant mouse, when approaching the area corresponding to the damaged glial wedge cell population, turned toward the ipsilateral lateral ventricle. This defect occurred before the appearance of ventriculomegaly. Discussion In conclusion, the abnormal development of the ventricular zone, which appears to be inherent to the etiology of several forms of congenital hydrocephalus, can explain, in some cases, the common association between hydrocephalus and corpus callosum dysgenesis. These results imply that further studies may be needed to understand the corpus callosum dysgenesis etiology when it concurs with hydrocephalus.
Collapse
Affiliation(s)
- Luis-Manuel Rodríguez-Pérez
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Universidad de Málaga, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
| | | | - Isabel de Diego
- Departamento de Anatomía y Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Malaga, Spain
| | - Aníbal Smith
- Departamento de Anatomía y Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Malaga, Spain
| | - Ruth Roales-Buján
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| | - Antonio J. Jiménez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| | - Patricia Paez-Gonzalez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Malaga, Spain
| |
Collapse
|
20
|
Cebul ER, Marivin A, Wexler LR, Perrat PN, Bénard CY, Garcia-Marcos M, Heiman MG. SAX-7/L1CAM acts with the adherens junction proteins MAGI-1, HMR-1/Cadherin, and AFD-1/Afadin to promote glial-mediated dendrite extension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575259. [PMID: 38260503 PMCID: PMC10802611 DOI: 10.1101/2024.01.11.575259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Adherens junctions (AJs) are a fundamental organizing structure for multicellular life. Although AJs are studied mainly in epithelia, their core function - stabilizing cell contacts by coupling adhesion molecules to the cytoskeleton - is important in diverse tissues. We find that two C. elegans sensory neurons, URX and BAG, require conserved AJ proteins for dendrite morphogenesis. We previously showed that URX and BAG dendrites attach to the embryonic nose via the adhesion molecule SAX-7/L1CAM, acting both in neurons and glia, and then extend by stretch during embryo elongation. Here, we find that a PDZ-binding motif (PB) in the SAX-7 cytoplasmic tail acts with other interaction motifs to promote dendrite extension. Using pull-down assays, we find that the SAX-7 PB binds the multi-PDZ scaffolding protein MAGI-1, which bridges it to the cadherin-catenin complex protein HMP-2/β-catenin. Using cell-specific rescue and depletion, we find that both MAGI-1 and HMR-1/Cadherin act in glia to non-autonomously promote dendrite extension. Double mutant analysis indicates that each protein can act independently of SAX-7, suggesting a multivalent adhesion complex. The SAX-7 PB motif also binds AFD-1/Afadin, loss of which further enhances sax-7 BAG dendrite defects. As MAGI-1, HMR-1, and AFD-1 are all found in epithelial AJs, we propose that an AJ-like complex in glia promotes dendrite extension.
Collapse
Affiliation(s)
- Elizabeth R. Cebul
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Present address: Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Arthur Marivin
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA, Boston University School of Medicine, Boston, MA 02118, USA
| | - Leland R. Wexler
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Paola N. Perrat
- Department of Biological Sciences, CERMO-FC Research Center, Universite du Québec à Montréal, Montreál, QC, Canada
| | - Claire Y. Bénard
- Department of Biological Sciences, CERMO-FC Research Center, Universite du Québec à Montréal, Montreál, QC, Canada
| | - Mikel Garcia-Marcos
- Department of Biochemistry & Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biology, College of Arts & Sciences, Boston University, Boston, MA 02115, USA
| | - Maxwell G. Heiman
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
21
|
Salmina AB, Alexandrova OP, Averchuk AS, Korsakova SA, Saridis MR, Illarioshkin SN, Yurchenko SO. Current progress and challenges in the development of brain tissue models: How to grow up the changeable brain in vitro? J Tissue Eng 2024; 15:20417314241235527. [PMID: 38516227 PMCID: PMC10956167 DOI: 10.1177/20417314241235527] [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: 09/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
In vitro modeling of brain tissue is a promising but not yet resolved problem in modern neurobiology and neuropharmacology. Complexity of the brain structure and diversity of cell-to-cell communication in (patho)physiological conditions make this task almost unachievable. However, establishment of novel in vitro brain models would ultimately lead to better understanding of development-associated or experience-driven brain plasticity, designing efficient approaches to restore aberrant brain functioning. The main goal of this review is to summarize the available data on methodological approaches that are currently in use, and to identify the most prospective trends in development of neurovascular unit, blood-brain barrier, blood-cerebrospinal fluid barrier, and neurogenic niche in vitro models. The manuscript focuses on the regulation of adult neurogenesis, cerebral microcirculation and fluids dynamics that should be reproduced in the in vitro 4D models to mimic brain development and its alterations in brain pathology. We discuss approaches that are critical for studying brain plasticity, deciphering the individual person-specific trajectory of brain development and aging, and testing new drug candidates in the in vitro models.
Collapse
Affiliation(s)
- Alla B Salmina
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Olga P Alexandrova
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Anton S Averchuk
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | | | | | | | | |
Collapse
|
22
|
Bear RM, Caspary T. Uncovering cilia function in glial development. Ann Hum Genet 2024; 88:27-44. [PMID: 37427745 PMCID: PMC10776815 DOI: 10.1111/ahg.12519] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023]
Abstract
Primary cilia play critical roles in regulating signaling pathways that underlie several developmental processes. In the nervous system, cilia are known to regulate signals that guide neuron development. Cilia dysregulation is implicated in neurological diseases, and the underlying mechanisms remain poorly understood. Cilia research has predominantly focused on neurons and has overlooked the diverse population of glial cells in the brain. Glial cells play essential roles during neurodevelopment, and their dysfunction contributes to neurological disease; however, the relationship between cilia function and glial development is understudied. Here we review the state of the field and highlight the glial cell types where cilia are found and the ciliary functions that are linked to glial development. This work uncovers the importance of cilia in glial development and raises outstanding questions for the field. We are poised to make progress in understanding the function of glial cilia in human development and their contribution to neurological diseases.
Collapse
Affiliation(s)
- Rachel M. Bear
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta GA 30322
- Graduate Program in Neuroscience
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta GA 30322
| |
Collapse
|
23
|
Sharma R, Rao M, Kanchan T, Pradhan S, Khera S. Incidental Subventricular Ependymal Cell Rests: A Rare Case Report. Am J Forensic Med Pathol 2023; 44:e120-e122. [PMID: 37669023 DOI: 10.1097/paf.0000000000000865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Affiliation(s)
| | | | - Tanuj Kanchan
- Forensic Medicine and Toxicology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | | | - Sudeep Khera
- From the Departments of Pathology & Lab Medicine
| |
Collapse
|
24
|
Wan Y, Holste KG, Ye F, Hua Y, Keep RF, Xi G. Minocycline attenuates hydrocephalus and inhibits iron accumulation, ependymal damage and epiplexus cell activation after intraventricular hemorrhage in aged rats. Exp Neurol 2023; 369:114523. [PMID: 37652293 PMCID: PMC10642526 DOI: 10.1016/j.expneurol.2023.114523] [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: 05/08/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Intracerebral hemorrhage is primarily a disease of the elderly and it is frequently accompanied by intraventricular hemorrhage (IVH) which can lead to posthemorrhagic hydrocephalus and poor prognosis. Red blood cell iron has been implicated in brain injury after cerebral hemorrhage. The current study examined using T2* magnetic resonance imaging (MRI) to detect periventricular iron deposition after IVH and investigated the effects of minocycline on hydrocephalus in an aged rat IVH model. It had three parts. In part 1, male aged rats received a 200 μl injection of saline or autologous blood into the lateral ventricle and were euthanized at day 14. In parts 2 and 3, aged IVH rats were treated with vehicle or minocycline and euthanized at day 7 or 14. Rats underwent MRI to quantify hydrocephalus and iron deposition followed by brain histology and immunohistochemistry. Periventricular iron overload was found after IVH using T2* MRI and confirmed by histology. IVH also caused ventricular wall damage and increased the number of CD68(+) choroid plexus epiplexus cells. Minocycline administration reduced iron deposition and ventricular volume at days 7 and 14 after IVH, as well as ventricle wall damage and epiplexus cell activation. In summary, IVH-induced hydrocephalus is associated with periventricular iron deposition, ependymal damage and choroid plexus epiplexus cell activation in aged rats. Minocycline attenuated those effects and might be a potential treatment for posthemorrhagic hydrocephalus in the elderly.
Collapse
Affiliation(s)
- Yingfeng Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Orrico-Sanchez A, Guiard BP, Manta S, Callebert J, Launay JM, Louis F, Paccard A, Gruszczynski C, Betancur C, Vialou V, Gautron S. Organic cation transporter 2 contributes to SSRI antidepressant efficacy by controlling tryptophan availability in the brain. Transl Psychiatry 2023; 13:302. [PMID: 37775532 PMCID: PMC10542329 DOI: 10.1038/s41398-023-02596-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRI) are common first-line treatments for major depression. However, a significant number of depressed patients do not respond adequately to these pharmacological treatments. In the present preclinical study, we demonstrate that organic cation transporter 2 (OCT2), an atypical monoamine transporter, contributes to the effects of SSRI by regulating the routing of the essential amino acid tryptophan to the brain. Contrarily to wild-type mice, OCT2-invalidated mice failed to respond to prolonged fluoxetine treatment in a chronic depression model induced by corticosterone exposure recapitulating core symptoms of depression, i.e., anhedonia, social withdrawal, anxiety, and memory impairment. After corticosterone and fluoxetine treatment, the levels of tryptophan and its metabolites serotonin and kynurenine were decreased in the brain of OCT2 mutant mice compared to wild-type mice and reciprocally tryptophan and kynurenine levels were increased in mutants' plasma. OCT2 was detected by immunofluorescence in several structures at the blood-cerebrospinal fluid (CSF) or brain-CSF interface. Tryptophan supplementation during fluoxetine treatment increased brain concentrations of tryptophan and, more discreetly, of 5-HT in wild-type and OCT2 mutant mice. Importantly, tryptophan supplementation improved the sensitivity to fluoxetine treatment of OCT2 mutant mice, impacting chiefly anhedonia and short-term memory. Western blot analysis showed that glycogen synthase kinase-3β (GSK3β) and mammalian/mechanistic target of rapamycin (mTOR) intracellular signaling was impaired in OCT2 mutant mice brain after corticosterone and fluoxetine treatment and, conversely, tryptophan supplementation recruited selectively the mTOR protein complex 2. This study provides the first evidence of the physiological relevance of OCT2-mediated tryptophan transport, and its biological consequences on serotonin homeostasis in the brain and SSRI efficacy.
Collapse
Affiliation(s)
| | - Bruno P Guiard
- Université Paul Sabatier, CNRS, Research Center on Animal Cognition, Toulouse, France
| | - Stella Manta
- Université Paul Sabatier, CNRS, Research Center on Animal Cognition, Toulouse, France
| | - Jacques Callebert
- Sorbonne Paris Cité, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean-Marie Launay
- Sorbonne Paris Cité, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Franck Louis
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | - Antoine Paccard
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | | | - Catalina Betancur
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France
| | - Vincent Vialou
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France.
| | - Sophie Gautron
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Paris, France.
| |
Collapse
|
26
|
Jackson RJ, Keiser MS, Meltzer JC, Fykstra DP, Dierksmeier SE, Melloni A, Nakajima T, Tecedor L, Ranum PT, Carrell E, Chen Y, Holtzman DM, Davidson BL, Hyman BT. APOE2 gene therapy reduces amyloid deposition, and improves markers of neuroinflammation and neurodegeneration in a mouse model of Alzheimer disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.552850. [PMID: 37645718 PMCID: PMC10461997 DOI: 10.1101/2023.08.14.552850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do they have a later age of onset, milder clinical course, and less severe neuropathological findings than others with Alzheimer disease. The contrast is especially stark in comparison to the phenotype associated with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, as well as a more aggressive clinical course and notably more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Even one APOE ε2 allele improves phenotype, but it is uncertain if that is due to the replacement of a more toxic allele by APOE ε2, or if APOE ε2 has a protective, neuro-modulatory effect. Here, we demonstrate that brain exposure to APOE2 via a gene therapy approach which bathes the entire cortical mantle in the gene product after transduction of the ependyma, rapidly ameliorates established Aβ plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE4. This result suggests a promising protective effect of exogenous APOE2, revealing a cell non-autonomous effect of the protein on microglial activation. We also show that plaque associated microglia in the brain of patients who inherit APOE2 similarly have less microglial reactivity to plaques. These data raise the potential that an APOE2 therapeutic could be effective in Alzheimer disease even in individuals born with the risk ε4 allele. One Sentence Summary Introduction of ApoE2 using an AAV that transduces the ependymal cells of the ventricle causes a reduction in amyloid load and plaque associated synapse loss, and reduces neuroinflammation by modulating microglial responsiveness to plaques.
Collapse
|
27
|
Nelles DG, Hazrati LN. The pathological potential of ependymal cells in mild traumatic brain injury. Front Cell Neurosci 2023; 17:1216420. [PMID: 37396927 PMCID: PMC10312375 DOI: 10.3389/fncel.2023.1216420] [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: 05/03/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is a common neurological condition affecting millions of individuals worldwide. Although the pathology of mTBI is not fully understood, ependymal cells present a promising approach for studying the pathogenesis of mTBI. Previous studies have revealed that DNA damage in the form of γH2AX accumulates in ependymal cells following mTBI, with evidence of widespread cellular senescence in the brain. Ependymal ciliary dysfunction has also been observed, leading to altered cerebrospinal fluid homeostasis. Even though ependymal cells have not been extensively studied in the context of mTBI, these observations reflect the pathological potential of ependymal cells that may underlie the neuropathological and clinical presentations of mTBI. This mini review explores the molecular and structural alterations that have been reported in ependymal cells following mTBI, as well as the potential pathological mechanisms mediated by ependymal cells that may contribute to overall dysfunction of the brain post-mTBI. Specifically, we address the topics of DNA damage-induced cellular senescence, dysregulation of cerebrospinal fluid homeostasis, and the consequences of impaired ependymal cell barriers. Moreover, we highlight potential ependymal cell-based therapies for the treatment of mTBI, with a focus on neurogenesis, ependymal cell repair, and modulation of senescence signaling pathways. Further insight and research in this field will help to establish the role of ependymal cells in the pathogenesis of mTBI and may lead to improved treatments that leverage ependymal cells to target the origins of mTBI pathology.
Collapse
Affiliation(s)
- Diana G. Nelles
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
28
|
Paez-Gonzalez P, Lopez-de-San-Sebastian J, Ceron-Funez R, Jimenez AJ, Rodríguez-Perez LM. Therapeutic strategies to recover ependymal barrier after inflammatory damage: relevance for recovering neurogenesis during development. Front Neurosci 2023; 17:1204197. [PMID: 37397456 PMCID: PMC10308384 DOI: 10.3389/fnins.2023.1204197] [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: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
The epithelium covering the surfaces of the cerebral ventricular system is known as the ependyma, and is essential for maintaining the physical and functional integrity of the central nervous system. Additionally, the ependyma plays an essential role in neurogenesis, neuroinflammatory modulation and neurodegenerative diseases. Ependyma barrier is severely affected by perinatal hemorrhages and infections that cross the blood brain barrier. The recovery and regeneration of ependyma after damage are key to stabilizing neuroinflammatory and neurodegenerative processes that are critical during early postnatal ages. Unfortunately, there are no effective therapies to regenerate this tissue in human patients. Here, the roles of the ependymal barrier in the context of neurogenesis and homeostasis are reviewed, and future research lines for development of actual therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Patricia Paez-Gonzalez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Raquel Ceron-Funez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
| | - Antonio J. Jimenez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
| | - Luis Manuel Rodríguez-Perez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Málaga, Spain
- Department of Human Physiology, Human Histology, Pathological Anatomy and Sports, University of Malaga, Málaga, Spain
| |
Collapse
|
29
|
Rive Le Gouard N, Nicolle R, Lefebvre M, Gelot A, Heide S, Gerasimenko A, Grigorescu R, Derive N, Jouannic JM, Garel C, Valence S, Quenum-Miraillet G, Chantot-Bastaraud S, Keren B, Heron D, Attie-Bitach T. First reports of fetal SMARCC1 related hydrocephalus. Eur J Med Genet 2023:104797. [PMID: 37285932 DOI: 10.1016/j.ejmg.2023.104797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/11/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023]
Abstract
The SMARCC1 gene has been involved in congenital ventriculomegaly with aqueduct stenosis but only a few patients have been reported so far, with no antenatal cases, and it is currently not annotated as a morbid gene in OMIM nor in the Human Phenotype Ontology. Most of the reported variants are loss of function (LoF) and are often inherited from unaffected parents. SMARCC1 encodes a subunit of the mSWI/SNF complex and affects the chromatin structure and expression of several genes. Here, we report the two first antenatal cases of SMARCC1 LoF variants detected by Whole Genome Sequencing (WGS). Ventriculomegaly is the common feature in those fetuses. Both identified variants are inherited from a healthy parent, which supports the reported incomplete penetrance of this gene. This makes the identification of this condition in WGS as well as the genetic counseling challenging.
Collapse
Affiliation(s)
- Nicolas Rive Le Gouard
- UF de Génomique du Développement, Département de Génétique médicale, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Paris, France.
| | - Romain Nicolle
- Service de Médecine Génomique des maladies rares, UF MP5, Hôpital Necker-Enfants Malades, AP-HP Université Paris Cité, Paris, France
| | | | - Antoinette Gelot
- Service de Foetopathologie, Hôpital Armand Trousseau, AP-HP Sorbonne Université, Paris, France
| | - Solveig Heide
- UF de Génétique Médicale et CRMR « Déficience intellectuelle », Département de Génétique médicale, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Paris, France
| | - Anna Gerasimenko
- UF de Génétique Médicale et CRMR « Déficience intellectuelle », Département de Génétique médicale, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Paris, France
| | - Romulus Grigorescu
- Service de Foetopathologie, Hôpital Armand Trousseau, AP-HP Sorbonne Université, Paris, France
| | - Nicolas Derive
- Laboratoire de Biologie Médicale Multisites SeqOIA, Paris, France
| | - Jean-Marie Jouannic
- Gynécologie obstétrique, Hôpital Trousseau, Centre de Référence C-MAVEM, AP-HP Sorbonne Université, Paris, France
| | - Catherine Garel
- Service de Radiologie Pédiatrique, Hôpital Trousseau, AP-HP Sorbonne Université, Paris, France
| | - Stéphanie Valence
- Service de Neurologie Pédiatrique, Hôpital Trousseau, AP-HP Sorbonne Université, Paris, France
| | - Geneviève Quenum-Miraillet
- UF de Génomique Chromosomique, Département de Génétique médicale, Hôpital Armand Trousseau, AP-HP Sorbonne Université, Paris, France
| | - Sandra Chantot-Bastaraud
- UF de Génomique Chromosomique, Département de Génétique médicale, Hôpital Armand Trousseau, AP-HP Sorbonne Université, Paris, France
| | - Boris Keren
- UF de Génomique du Développement, Département de Génétique médicale, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Paris, France; Laboratoire de Biologie Médicale Multisites SeqOIA, Paris, France
| | - Delphine Heron
- UF de Génétique Médicale et CRMR « Déficience intellectuelle », Département de Génétique médicale, Groupe Hospitalier Pitié-Salpêtrière, AP-HP Sorbonne Université, Paris, France
| | - Tania Attie-Bitach
- Service de Médecine Génomique des maladies rares, UF MP5, Hôpital Necker-Enfants Malades, AP-HP Université Paris Cité, Paris, France; Laboratoire de Biologie Médicale Multisites SeqOIA, Paris, France.
| |
Collapse
|
30
|
Ma XY, Yang TT, Liu L, Peng XC, Qian F, Tang FR. Ependyma in Neurodegenerative Diseases, Radiation-Induced Brain Injury and as a Therapeutic Target for Neurotrophic Factors. Biomolecules 2023; 13:754. [PMID: 37238624 PMCID: PMC10216700 DOI: 10.3390/biom13050754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The neuron loss caused by the progressive damage to the nervous system is proposed to be the main pathogenesis of neurodegenerative diseases. Ependyma is a layer of ciliated ependymal cells that participates in the formation of the brain-cerebrospinal fluid barrier (BCB). It functions to promotes the circulation of cerebrospinal fluid (CSF) and the material exchange between CSF and brain interstitial fluid. Radiation-induced brain injury (RIBI) shows obvious impairments of the blood-brain barrier (BBB). In the neuroinflammatory processes after acute brain injury, a large amount of complement proteins and infiltrated immune cells are circulated in the CSF to resist brain damage and promote substance exchange through the BCB. However, as the protective barrier lining the brain ventricles, the ependyma is extremely vulnerable to cytotoxic and cytolytic immune responses. When the ependyma is damaged, the integrity of BCB is destroyed, and the CSF flow and material exchange is affected, leading to brain microenvironment imbalance, which plays a vital role in the pathogenesis of neurodegenerative diseases. Epidermal growth factor (EGF) and other neurotrophic factors promote the differentiation and maturation of ependymal cells to maintain the integrity of the ependyma and the activity of ependymal cilia, and may have therapeutic potential in restoring the homeostasis of the brain microenvironment after RIBI or during the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin-Yu Ma
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Ting-Ting Yang
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Lian Liu
- Department of Pharmacology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
31
|
Modvig S, Jeyakumar J, Marquart HV, Christensen C. Integrins and the Metastasis-like Dissemination of Acute Lymphoblastic Leukemia to the Central Nervous System. Cancers (Basel) 2023; 15:cancers15092504. [PMID: 37173970 PMCID: PMC10177281 DOI: 10.3390/cancers15092504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) disseminates with high prevalence to the central nervous system (CNS) in a process resembling aspects of the CNS surveillance of normal immune cells as well as aspects of brain metastasis from solid cancers. Importantly, inside the CNS, the ALL blasts are typically confined within the cerebrospinal fluid (CSF)-filled cavities of the subarachnoid space, which they use as a sanctuary protected from both chemotherapy and immune cells. At present, high cumulative doses of intrathecal chemotherapy are administered to patients, but this is associated with neurotoxicity and CNS relapse still occurs. Thus, it is imperative to identify markers and novel therapy targets specific to CNS ALL. Integrins represent a family of adhesion molecules involved in cell-cell and cell-matrix interactions, implicated in the adhesion and migration of metastatic cancer cells, normal immune cells, and leukemic blasts. The ability of integrins to also facilitate cell-adhesion mediated drug resistance, combined with recent discoveries of integrin-dependent routes of leukemic cells into the CNS, have sparked a renewed interest in integrins as markers and therapeutic targets in CNS leukemia. Here, we review the roles of integrins in CNS surveillance by normal lymphocytes, dissemination to the CNS by ALL cells, and brain metastasis from solid cancers. Furthermore, we discuss whether ALL dissemination to the CNS abides by known hallmarks of metastasis, and the potential roles of integrins in this context.
Collapse
Affiliation(s)
- Signe Modvig
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jenani Jeyakumar
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Claus Christensen
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
32
|
Verkhratsky A, Pivoriūnas A. Astroglia support, regulate and reinforce brain barriers. Neurobiol Dis 2023; 179:106054. [PMID: 36842485 DOI: 10.1016/j.nbd.2023.106054] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/28/2023] Open
Abstract
Nervous system is segregated from the body by the complex system of barriers. The CNS is protected by (i) the blood-brain and blood-spinal cord barrier between the intracerebral and intraspinal blood vessels and the brain parenchyma; (ii) the arachnoid blood-cerebrospinal fluid barrier; (iii) the blood-cerebrospinal barrier of circumventricular organs made by tanycytes and (iv) the choroid plexus blood-CSF barrier formed by choroid ependymocytes. In the peripheral nervous system the nerve-blood barrier is secured by tight junctions between specialised glial cells known as perineural cells. In the CNS astroglia contribute to all barriers through the glia limitans, which represent the parenchymal portion of the barrier system. Astroglia through secretion of various paracrine factors regulate the permeability of endothelial vascular barrier; in pathology damage or asthenia of astrocytes may compromise brain barriers integrity.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| |
Collapse
|
33
|
García-Bonilla M, Ojeda-Pérez B, Shumilov K, Rodríguez-Pérez LM, Domínguez-Pinos D, Vitorica J, Jiménez S, Ramírez-Lorca R, Echevarría M, Cárdenas-García C, Iglesias T, Gutiérrez A, McAllister JP, Limbrick DD, Páez-González P, Jiménez AJ. Generation of Periventricular Reactive Astrocytes Overexpressing Aquaporin 4 Is Stimulated by Mesenchymal Stem Cell Therapy. Int J Mol Sci 2023; 24:5640. [PMID: 36982724 PMCID: PMC10057840 DOI: 10.3390/ijms24065640] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Aquaporin-4 (AQP4) plays a crucial role in brain water circulation and is considered a therapeutic target in hydrocephalus. Congenital hydrocephalus is associated with a reaction of astrocytes in the periventricular white matter both in experimental models and human cases. A previous report showed that bone marrow-derived mesenchymal stem cells (BM-MSCs) transplanted into the lateral ventricles of hyh mice exhibiting severe congenital hydrocephalus are attracted by the periventricular astrocyte reaction, and the cerebral tissue displays recovery. The present investigation aimed to test the effect of BM-MSC treatment on astrocyte reaction formation. BM-MSCs were injected into the lateral ventricles of four-day-old hyh mice, and the periventricular reaction was detected two weeks later. A protein expression analysis of the cerebral tissue differentiated the BM-MSC-treated mice from the controls and revealed effects on neural development. In in vivo and in vitro experiments, BM-MSCs stimulated the generation of periventricular reactive astrocytes overexpressing AQP4 and its regulatory protein kinase D-interacting substrate of 220 kDa (Kidins220). In the cerebral tissue, mRNA overexpression of nerve growth factor (NGF), vascular endothelial growth factor (VEGF), hypoxia-inducible factor-1 (HIF1α), and transforming growth factor beta 1 (TGFβ1) could be related to the regulation of the astrocyte reaction and AQP4 expression. In conclusion, BM-MSC treatment in hydrocephalus can stimulate a key developmental process such as the periventricular astrocyte reaction, where AQP4 overexpression could be implicated in tissue recovery.
Collapse
Affiliation(s)
- María García-Bonilla
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| | - Betsaida Ojeda-Pérez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| | - Kirill Shumilov
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Luis-Manuel Rodríguez-Pérez
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, University of Malaga, 29010 Malaga, Spain
| | | | - Javier Vitorica
- Department of Molecular Biology and Biochemistry, University of Seville, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
| | - Sebastián Jiménez
- Department of Molecular Biology and Biochemistry, University of Seville, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
| | - Reposo Ramírez-Lorca
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
- Department of Physiology and Biophysics, University of Seville, 41009 Seville, Spain
| | - Miriam Echevarría
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
- Department of Physiology and Biophysics, University of Seville, 41009 Seville, Spain
| | - Casimiro Cárdenas-García
- Servicios Centrales de Apoyo a la Investigación (SCAI), University of Malaga, 29010 Malaga, Spain
| | - Teresa Iglesias
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain
| | - Antonia Gutiérrez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
| | - James P. McAllister
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - David D. Limbrick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Patricia Páez-González
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| | - Antonio J. Jiménez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| |
Collapse
|
34
|
Karimy JK, Newville JC, Sadegh C, Morris JA, Monuki ES, Limbrick DD, McAllister Ii JP, Koschnitzky JE, Lehtinen MK, Jantzie LL. Outcomes of the 2019 hydrocephalus association workshop, "Driving common pathways: extending insights from posthemorrhagic hydrocephalus". Fluids Barriers CNS 2023; 20:4. [PMID: 36639792 PMCID: PMC9838022 DOI: 10.1186/s12987-023-00406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The Hydrocephalus Association (HA) workshop, Driving Common Pathways: Extending Insights from Posthemorrhagic Hydrocephalus, was held on November 4 and 5, 2019 at Washington University in St. Louis. The workshop brought together a diverse group of basic, translational, and clinical scientists conducting research on multiple hydrocephalus etiologies with select outside researchers. The main goals of the workshop were to explore areas of potential overlap between hydrocephalus etiologies and identify drug targets that could positively impact various forms of hydrocephalus. This report details the major themes of the workshop and the research presented on three cell types that are targets for new hydrocephalus interventions: choroid plexus epithelial cells, ventricular ependymal cells, and immune cells (macrophages and microglia).
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Family Medicine, Mountain Area Health Education Center - Boone, North Carolina, 28607, USA
| | - Jessie C Newville
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Cameron Sadegh
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, MA, Boston, 02114, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Jill A Morris
- National Institute of Neurological Disorders and Stroke, Neuroscience Center, National Institutes of Health, 6001 Executive Blvd, NSC Rm 2112, Bethesda, MD, 20892, USA
| | - Edwin S Monuki
- Departments of Pathology & Laboratory Medicine and Developmental & Cell Biology, University of California Irvine, Irvine, CA, 92697, USA
| | - David D Limbrick
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - James P McAllister Ii
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | | | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Lauren L Jantzie
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
- Kennedy Krieger Institute, Baltimore, MD, 21287, USA.
| |
Collapse
|
35
|
Lopez-Rodriguez D, Rohrbach A, Lanzillo M, Gervais M, Croizier S, Langlet F. Ontogeny of ependymoglial cells lining the third ventricle in mice. Front Endocrinol (Lausanne) 2023; 13:1073759. [PMID: 36686420 PMCID: PMC9849764 DOI: 10.3389/fendo.2022.1073759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/02/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction During hypothalamic development, the germinative neuroepithelium gives birth to diverse neural cells that regulate numerous physiological functions in adulthood. Methods Here, we studied the ontogeny of ependymal cells in the mouse mediobasal hypothalamus using the BrdU approach and publicly available single-cell RNAseq datasets. Results We observed that while typical ependymal cells are mainly produced at E13, tanycyte birth depends on time and subtypes and lasts up to P8. Typical ependymocytes and β tanycytes are the first to arise at the top and bottom of the dorsoventral axis around E13, whereas α tanycytes emerge later in development, generating an outside-in dorsoventral gradient along the third ventricle. Additionally, α tanycyte generation displayed a rostral-to-caudal pattern. Finally, tanycytes mature progressively until they reach transcriptional maturity between P4 and P14. Discussion Altogether, this data shows that ependyma generation differs in time and distribution, highlighting the heterogeneity of the third ventricle.
Collapse
Affiliation(s)
- David Lopez-Rodriguez
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antoine Rohrbach
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Marc Lanzillo
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Manon Gervais
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
36
|
Bigbee JW. Cells of the Central Nervous System: An Overview of Their Structure and Function. ADVANCES IN NEUROBIOLOGY 2023; 29:41-64. [PMID: 36255671 DOI: 10.1007/978-3-031-12390-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The central nervous system is the last major organ system in the vertebrate body to yield its cellular structure, due to the complexity of its cells and their interactions. The fundamental unit of the nervous system is the neuron, which forms complex circuits that receive and integrate information and generate adaptive responses. Each neuron is composed of an input domain consisting of multiple dendrites along with the cell body, which is also responsible for the majority of macromolecule synthesis for the cell. The output domain is the axon which is a singular extension from the cell body that propagates the action potential to the synapse, where signals pass from one neuron to another. Facilitating these functions are cohorts of supporting cells consisting of astrocytes, oligodendrocytes and microglia along with NG2 cells and ependymal cells. Astrocytes have a dazzling array of functions including physical support, maintenance of homeostasis, development and integration of synaptic activity. Oligodendrocytes form the myelin sheath which surrounds axons and enables rapid conduction of the nerve impulse. Microglia are the resident immune cells, providing immune surveillance and remodeling of neuronal circuits during development and trauma. All these cells function in concert with each other, producing the remarkably diverse functions of the nervous system.
Collapse
Affiliation(s)
- John W Bigbee
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
37
|
D'Gama PP, Jurisch-Yaksi N. Methods to study motile ciliated cell types in the zebrafish brain. Methods Cell Biol 2023; 176:103-123. [PMID: 37164533 DOI: 10.1016/bs.mcb.2023.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Cilia are well conserved hair-like structures that have diverse sensory and motile functions. In the brain, motile ciliated cells, known as ependymal cells, line the cerebrospinal fluid (CSF) filled ventricles, where their beating contribute to fluid movement. Ependymal cells have gathered increasing interest since they are associated with hydrocephalus, a neurological condition with ventricular enlargement. In this article, we highlight methods to identify and characterize motile ciliated ependymal lineage in the brain of zebrafish using histological staining and transgenic reporter lines.
Collapse
|
38
|
Nelles DG, Hazrati LN. Ependymal cells and neurodegenerative disease: outcomes of compromised ependymal barrier function. Brain Commun 2022; 4:fcac288. [PMID: 36415662 PMCID: PMC9677497 DOI: 10.1093/braincomms/fcac288] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/13/2022] [Accepted: 11/01/2022] [Indexed: 08/08/2023] Open
Abstract
Within the central nervous system, ependymal cells form critical components of the blood-cerebrospinal fluid barrier and the cerebrospinal fluid-brain barrier. These barriers provide biochemical, immunological and physical protection against the entry of molecules and foreign substances into the cerebrospinal fluid while also regulating cerebrospinal fluid dynamics, such as the composition, flow and removal of waste from the cerebrospinal fluid. Previous research has demonstrated that several neurodegenerative diseases, such as Alzheimer's disease and multiple sclerosis, display irregularities in ependymal cell function, morphology, gene expression and metabolism. Despite playing key roles in maintaining overall brain health, ependymal barriers are largely overlooked and understudied in the context of disease, thus limiting the development of novel diagnostic and treatment options. Therefore, this review explores the anatomical properties, functions and structures that define ependymal cells in the healthy brain, as well as the ways in which ependymal cell dysregulation manifests across several neurodegenerative diseases. Specifically, we will address potential mechanisms, causes and consequences of ependymal cell dysfunction and describe how compromising the integrity of ependymal barriers may initiate, contribute to, or drive widespread neurodegeneration in the brain.
Collapse
Affiliation(s)
- Diana G Nelles
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, 555 University Ave, Canada
| | - Lili-Naz Hazrati
- Correspondence to: Dr. Lili-Naz Hazrati 555 University Ave, Toronto ON M5G 1X8, Canada E-mail:
| |
Collapse
|
39
|
Caçoilo A, Rusinek H, Weickenmeier J. 3D finite-element brain modeling of lateral ventricular wall loading to rationalize periventricular white matter hyperintensity locations. ENGINEERING WITH COMPUTERS 2022; 38:3939-3955. [PMID: 37485473 PMCID: PMC10361695 DOI: 10.1007/s00366-022-01700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/19/2022] [Indexed: 07/25/2023]
Abstract
Aging-related periventricular white matter hyperintensities (pvWMHs) are a common observation in medical images of the aging brain. The underlying tissue damage is part of the complex pathophysiology associated with age-related microstructural changes and cognitive decline. PvWMH formation is linked to blood-brain barrier dysfunction from cerebral small vessel disease as well as the accumulation of cerebrospinal fluid in periventricular tissue due to progressive denudation of the ventricular wall. In need of a unifying theory for pvWMH etiology, image-based finite-element modeling is used to demonstrate that ventricular expansion from age-related cerebral atrophy and hemodynamic loading leads to maximum mechanical loading of the ventricular wall in the same locations that show pvWMHs. Ventricular inflation, induced via pressurization of the ventricular wall, creates significant ventricular wall stretch and stress on the ependymal cells lining the wall, that are linked to cerebrospinal fluid leaking from the lateral ventricles into periventricular white matter tissue. Eight anatomically accurate 3D brain models of cognitively healthy subjects with a wide range of ventricular shapes are created. For all models, our simulations show that mechanomarkers of mechanical wall loading are consistently highest in pvWMHs locations (p < 0.05). Maximum principal strain, the ependymal cell thinning ratio, and wall curvature are on average 14%, 8%, and 24% higher in pvWMH regions compared to the remaining ventricular wall, respectively. Computational modeling provides a powerful framework to systematically study pvWMH formation and growth with the goal to develop pharmacological interventions in the future.
Collapse
Affiliation(s)
- Andreia Caçoilo
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Henry Rusinek
- Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
40
|
Yoshida K, Chambers JK, Uchida K. Systemic Streptococcus agalactiae infection with meningo-ventriculitis in a Linnaeus's two-toed sloth (Choloepus didactylus). J Vet Med Sci 2022; 84:1417-1421. [PMID: 36058878 DOI: 10.1292/jvms.22-0317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A captive male Linnaeus's two-toed sloth died without any obvious clinical signs. At necropsy, multifocal ulceration at the lumbar and perianal skin, mitral valve vegetation, and multifocal hemorrhage in the leptomeninges were observed. Histopathologically, suppurative meningo-ventriculitis, dermatitis, and endocarditis characterized by severe neutrophilic infiltration were observed. Gram-positive cocci arranged in pairs or chains were present in these inflammatory lesions. Streptococcus agalactiae gene was detected in the skin, heart, and brain tissues by PCR and sequence analysis. These findings may indicate that S. agalactiae primarily infected the skin and then caused septicemia resulting in endocarditis and meningo-ventriculitis. The present case suggests that S. agalactiae infection can cause severe meningo-ventriculitis in two-toed sloth without any specific clinical signs.
Collapse
Affiliation(s)
- Kio Yoshida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - James K Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Harkins D, Harvey TJ, Atterton C, Miller I, Currey L, Oishi S, Kasherman M, Davila RA, Harris L, Green K, Piper H, Parton RG, Thor S, Cooper HM, Piper M. Hydrocephalus in Nfix−/− Mice Is Underpinned by Changes in Ependymal Cell Physiology. Cells 2022; 11:cells11152377. [PMID: 35954220 PMCID: PMC9368351 DOI: 10.3390/cells11152377] [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: 07/13/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
Nuclear factor one X (NFIX) is a transcription factor required for normal ependymal development. Constitutive loss of Nfix in mice (Nfix−/−) is associated with hydrocephalus and sloughing of the dorsal ependyma within the lateral ventricles. Previous studies have implicated NFIX in the transcriptional regulation of genes encoding for factors essential to ependymal development. However, the cellular and molecular mechanisms underpinning hydrocephalus in Nfix−/− mice are unknown. To investigate the role of NFIX in hydrocephalus, we examined ependymal cells in brains from postnatal Nfix−/− and control (Nfix+/+) mice using a combination of confocal and electron microscopy. This revealed that the ependymal cells in Nfix−/− mice exhibited abnormal cilia structure and disrupted localisation of adhesion proteins. Furthermore, we modelled ependymal cell adhesion using epithelial cell culture and revealed changes in extracellular matrix and adherens junction gene expression following knockdown of NFIX. Finally, the ablation of Nfix from ependymal cells in the adult brain using a conditional approach culminated in enlarged ventricles, sloughing of ependymal cells from the lateral ventricles and abnormal localisation of adhesion proteins, which are phenotypes observed during development. Collectively, these data demonstrate a pivotal role for NFIX in the regulation of cell adhesion within ependymal cells of the lateral ventricles.
Collapse
Affiliation(s)
- Danyon Harkins
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Tracey J. Harvey
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Cooper Atterton
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Ingrid Miller
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Laura Currey
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Sabrina Oishi
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Maria Kasherman
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Raul Ayala Davila
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Lucy Harris
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane 4072, Australia; (L.H.); (K.G.); (R.G.P.)
| | - Kathryn Green
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane 4072, Australia; (L.H.); (K.G.); (R.G.P.)
| | - Hannah Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Robert G. Parton
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane 4072, Australia; (L.H.); (K.G.); (R.G.P.)
- Institute for Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Stefan Thor
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
| | - Helen M. Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia;
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; (D.H.); (T.J.H.); (C.A.); (I.M.); (L.C.); (S.O.); (M.K.); (R.A.D.); (H.P.); (S.T.)
- Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia;
- Correspondence:
| |
Collapse
|
42
|
Abstract
Understanding normal brain aging physiology is essential to improving healthy human longevity, differentiation, and early detection of diseases, such as neurodegenerative diseases, which are an enormous social and economic burden. Functional decline, such as reduced physical activity and cognitive abilities, is typically associated with brain aging. The authors summarize the aging brain mechanism and effects of aging on the brain observed by brain structural MR imaging and advanced neuroimaging techniques, such as diffusion tensor imaging and functional MR imaging.
Collapse
Affiliation(s)
- Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 East Medical Center Drive, UH B2, Ann Arbor, MI 48109, USA
| | - Gaurang Shah
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 East Medical Center Drive, UH B2, Ann Arbor, MI 48109, USA.
| |
Collapse
|
43
|
Ji W, Tang Z, Chen Y, Wang C, Tan C, Liao J, Tong L, Xiao G. Ependymal Cilia: Physiology and Role in Hydrocephalus. Front Mol Neurosci 2022; 15:927479. [PMID: 35903173 PMCID: PMC9315228 DOI: 10.3389/fnmol.2022.927479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/20/2022] [Indexed: 01/10/2023] Open
Abstract
Cerebrospinal fluid (CSF), a colorless liquid that generally circulates from the lateral ventricles to the third and fourth ventricles, provides essential nutrients for brain homeostasis and growth factors during development. As evidenced by an increasing corpus of research, CSF serves a range of important functions. While it is considered that decreased CSF flow is associated to the development of hydrocephalus, it has recently been postulated that motile cilia, which line the apical surfaces of ependymal cells (ECs), play a role in stimulating CSF circulation by cilia beating. Ependymal cilia protrude from ECs, and their synchronous pulsing transports CSF from the lateral ventricle to the third and fourth ventricles, and then to the subarachnoid cavity for absorption. As a result, we postulated that malfunctioning ependymal cilia could disrupt normal CSF flow, raising the risk of hydrocephalus. This review aims to demonstrate the physiological functions of ependymal cilia, as well as how cilia immobility or disorientation causes problems. We also conclude conceivable ways of treatment of hydrocephalus currently for clinical application and provide theoretical support for regimen improvements by investigating the relationship between ependymal cilia and hydrocephalus development.
Collapse
Affiliation(s)
- Weiye Ji
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yibing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chuansen Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Changwu Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Tong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Gelei Xiao,
| |
Collapse
|
44
|
Norton ES, Whaley LA, Ulloa-Navas MJ, García-Tárraga P, Meneses KM, Lara-Velazquez M, Zarco N, Carrano A, Quiñones-Hinojosa A, García-Verdugo JM, Guerrero-Cázares H. Glioblastoma disrupts the ependymal wall and extracellular matrix structures of the subventricular zone. Fluids Barriers CNS 2022; 19:58. [PMID: 35821139 PMCID: PMC9277938 DOI: 10.1186/s12987-022-00354-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive and common type of primary brain tumor in adults. Tumor location plays a role in patient prognosis, with tumors proximal to the lateral ventricles (LVs) presenting with worse overall survival, increased expression of stem cell genes, and increased incidence of distal tumor recurrence. This may be due in part to interaction of GBM with factors of the subventricular zone (SVZ), including those contained within the cerebrospinal fluid (CSF). However, direct interaction of GBM tumors with CSF has not been proved and would be hindered in the presence of an intact ependymal cell layer. Methods Here, we investigate the ependymal cell barrier and its derived extracellular matrix (ECM) fractones in the vicinity of a GBM tumor. Patient-derived GBM cells were orthotopically implanted into immunosuppressed athymic mice in locations distal and proximal to the LV. A PBS vehicle injection in the proximal location was included as a control. At four weeks post-xenograft, brain tissue was examined for alterations in ependymal cell health via immunohistochemistry, scanning electron microscopy, and transmission electron microscopy. Results We identified local invading GBM cells within the LV wall and increased influx of CSF into the LV-proximal GBM tumor bulk compared to controls. In addition to the physical disruption of the ependymal cell barrier, we also identified increased signs of compromised ependymal cell health in LV-proximal tumor-bearing mice. These signs include increased accumulation of lipid droplets, decreased cilia length and number, and decreased expression of cell channel proteins. We additionally identified elevated numbers of small fractones in the SVZ within this group, suggesting increased indirect CSF-contained molecule signaling to tumor cells. Conclusions Our data is the first to show that LV-proximal GBMs physically disrupt the ependymal cell barrier in animal models, resulting in disruptions in ependymal cell biology and increased CSF interaction with the tumor bulk. These findings point to ependymal cell health and CSF-contained molecules as potential axes for therapeutic targeting in the treatment of GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00354-8.
Collapse
Affiliation(s)
- Emily S Norton
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Lauren A Whaley
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Department of Biology, University of North Florida, Jacksonville, FL, USA
| | - María José Ulloa-Navas
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Patricia García-Tárraga
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Kayleah M Meneses
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Hugo Guerrero-Cázares
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
45
|
Abstract
The brain harbors a unique ability to, figuratively speaking, shift its gears. During wakefulness, the brain is geared fully toward processing information and behaving, while homeostatic functions predominate during sleep. The blood-brain barrier establishes a stable environment that is optimal for neuronal function, yet the barrier imposes a physiological problem; transcapillary filtration that forms extracellular fluid in other organs is reduced to a minimum in brain. Consequently, the brain depends on a special fluid [the cerebrospinal fluid (CSF)] that is flushed into brain along the unique perivascular spaces created by astrocytic vascular endfeet. We describe this pathway, coined the term glymphatic system, based on its dependency on astrocytic vascular endfeet and their adluminal expression of aquaporin-4 water channels facing toward CSF-filled perivascular spaces. Glymphatic clearance of potentially harmful metabolic or protein waste products, such as amyloid-β, is primarily active during sleep, when its physiological drivers, the cardiac cycle, respiration, and slow vasomotion, together efficiently propel CSF inflow along periarterial spaces. The brain's extracellular space contains an abundance of proteoglycans and hyaluronan, which provide a low-resistance hydraulic conduit that rapidly can expand and shrink during the sleep-wake cycle. We describe this unique fluid system of the brain, which meets the brain's requisites to maintain homeostasis similar to peripheral organs, considering the blood-brain-barrier and the paths for formation and egress of the CSF.
Collapse
Affiliation(s)
- Martin Kaag Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
46
|
Garcia-Bonilla M, Castaneyra-Ruiz L, Zwick S, Talcott M, Otun A, Isaacs AM, Morales DM, Limbrick DD, McAllister JP. Acquired hydrocephalus is associated with neuroinflammation, progenitor loss, and cellular changes in the subventricular zone and periventricular white matter. Fluids Barriers CNS 2022; 19:17. [PMID: 35193620 PMCID: PMC8864805 DOI: 10.1186/s12987-022-00313-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/06/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Hydrocephalus is a neurological disease with an incidence of 80-125 per 100,000 births in the United States. Neuropathology comprises ventriculomegaly, periventricular white matter (PVWM) alterations, inflammation, and gliosis. We hypothesized that hydrocephalus in a pig model is associated with subventricular and PVWM cellular alterations and neuroinflammation that could mimic the neuropathology described in hydrocephalic infants. METHODS Hydrocephalus was induced by intracisternal kaolin injections in 35-day old female pigs (n = 7 for tissue analysis, n = 10 for CSF analysis). Age-matched sham controls received saline injections (n = 6). After 19-40 days, MRI scanning was performed to measure the ventricular volume. Stem cell proliferation was studied in the Subventricular Zone (SVZ), and cell death and oligodendrocytes were examined in the PVWM. The neuroinflammatory reaction was studied by quantifying astrocytes and microglial cells in the PVWM, and inflammatory cytokines in the CSF. RESULTS The expansion of the ventricles was especially pronounced in the body of the lateral ventricle, where ependymal disruption occurred. PVWM showed a 44% increase in cell death and a 67% reduction of oligodendrocytes. In the SVZ, the number of proliferative cells and oligodendrocyte decreased by 75% and 57% respectively. The decrease of the SVZ area correlated significantly with ventricular volume increase. Neuroinflammation occurred in the hydrocephalic pigs with a significant increase of astrocytes and microglia in the PVWM, and high levels of inflammatory interleukins IL-6 and IL-8 in the CSF. CONCLUSION The induction of acquired hydrocephalus produced alterations in the PVWM, reduced cell proliferation in the SVZ, and neuroinflammation.
Collapse
Affiliation(s)
- Maria Garcia-Bonilla
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA.
| | - Leandro Castaneyra-Ruiz
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Sarah Zwick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Michael Talcott
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA.,Division of Comparative Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Ayodamola Otun
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Albert M Isaacs
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Calgary, Alberta, T2N 2T9, Canada
| | - Diego M Morales
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - James P McAllister
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
47
|
Central Nervous System Stimulants Limit Caffeine Transport at the Blood-Cerebrospinal Fluid Barrier. Int J Mol Sci 2022; 23:ijms23031862. [PMID: 35163784 PMCID: PMC8836437 DOI: 10.3390/ijms23031862] [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: 01/05/2022] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 12/10/2022] Open
Abstract
Caffeine, a common ingredient in energy drinks, crosses the blood-brain barrier easily, but the kinetics of caffeine across the blood-cerebrospinal fluid barrier (BCSFB) has not been investigated. Therefore, 127 autopsy cases (Group A, 30 patients, stimulant-detected group; and Group B, 97 patients, no stimulant detected group) were examined. In addition, a BCSFB model was constructed using human vascular endothelial cells and human choroid plexus epithelial cells separated by a filter, and the kinetics of caffeine in the BCSFB and the effects of 4-aminopyridine (4-AP), a neuroexcitatory agent, were studied. Caffeine concentrations in right heart blood (Rs) and cerebrospinal fluid (CSF) were compared in the autopsy cases: caffeine concentrations were higher in Rs than CSF in Group A compared to Group B. In the BCSFB model, caffeine and 4-AP were added to the upper layer, and the concentration in the lower layer of choroid plexus epithelial cells was measured. The CSF caffeine concentration was suppressed, depending on the 4-AP concentration. Histomorphological examination suggested that choroid plexus epithelial cells were involved in inhibiting the efflux of caffeine to the CSF. Thus, the simultaneous presence of stimulants and caffeine inhibits caffeine transfer across the BCSFB.
Collapse
|
48
|
Cooze BJ, Dickerson M, Loganathan R, Watkins LM, Grounds E, Pearson BR, Bevan RJ, Morgan BP, Magliozzi R, Reynolds R, Neal JW, Howell OW. The association between neurodegeneration and local complement activation in the thalamus to progressive multiple sclerosis outcome. Brain Pathol 2022; 32:e13054. [PMID: 35132719 PMCID: PMC9425007 DOI: 10.1111/bpa.13054] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/17/2021] [Accepted: 01/17/2022] [Indexed: 01/22/2023] Open
Abstract
The extent of grey matter demyelination and neurodegeneration in the progressive multiple sclerosis (PMS) brains at post‐mortem associates with more severe disease. Regional tissue atrophy, especially affecting the cortical and deep grey matter, including the thalamus, is prognostic for poor outcomes. Microglial and complement activation are important in the pathogenesis and contribute to damaging processes that underlie tissue atrophy in PMS. We investigated the extent of pathology and innate immune activation in the thalamus in comparison to cortical grey and white matter in blocks from 21 cases of PMS and 10 matched controls. Using a digital pathology workflow, we show that the thalamus is invariably affected by demyelination and had a far higher proportion of active inflammatory lesions than forebrain cortical tissue blocks from the same cases. Lesions were larger and more frequent in the medial nuclei near the ventricular margin, whilst neuronal loss was greatest in the lateral thalamic nuclei. The extent of thalamic neuron loss was not associated with thalamic demyelination but correlated with the burden of white matter pathology in other forebrain areas (Spearman r = 0.79, p < 0.0001). Only thalamic neuronal loss, and not that seen in other forebrain cortical areas, correlated with disease duration (Spearman r = −0.58, p = 0.009) and age of death (Spearman r = −0.47, p = 0.045). Immunoreactivity for the complement pattern recognition molecule C1q, and products of complement activation (C4d, Bb and C3b) were elevated in thalamic lesions with an active inflammatory pathology. Complement regulatory protein, C1 inhibitor, was unchanged in expression. We conclude that active inflammatory demyelination, neuronal loss and local complement synthesis and activation in the thalamus, are important to the pathological and clinical disease outcomes of PMS.
Collapse
Affiliation(s)
- Benjamin J Cooze
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Matthew Dickerson
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | | | - Lewis M Watkins
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ethan Grounds
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ben R Pearson
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ryan Jack Bevan
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - Roberta Magliozzi
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | | | - James W Neal
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Owain W Howell
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| |
Collapse
|
49
|
Targeting nanoparticles to malignant tumors. Biochim Biophys Acta Rev Cancer 2022; 1877:188703. [DOI: 10.1016/j.bbcan.2022.188703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/01/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022]
|
50
|
Phosphorylation-dependent proteome of Marcks in ependyma during aging and behavioral homeostasis in the mouse forebrain. GeroScience 2022; 44:2077-2094. [DOI: 10.1007/s11357-022-00517-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 01/13/2022] [Indexed: 11/04/2022] Open
|