1
|
Pereira M, Droguerre M, Valdebenito M, Vidal L, Marcy G, Benkeder S, Marchal P, Comte JC, Pascual O, Zimmer L, Vidal B. Induction of haemodynamic travelling waves by glial-related vasomotion in a rat model of neuroinflammation: implications for functional neuroimaging. EBioMedicine 2025; 116:105777. [PMID: 40435721 PMCID: PMC12159494 DOI: 10.1016/j.ebiom.2025.105777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 05/05/2025] [Accepted: 05/14/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Cerebral haemodynamics are crucial for brain homoeostasis and serve as a key proxy for brain activity. Although this process involves coordinated interaction between vessels, neurons, and glial cells, its dysregulation in neuroinflammation is not well understood. METHODS We used in vivo mesoscopic functional ultrasound imaging to monitor cerebral blood volume changes during neuroinflammation in male rats injected with lipopolysaccharide (LPS) in the visual cortex, under resting-state or visual stimulation, combined to advanced ex vivo techniques for glial cell reactivity analysis. FINDINGS Cortical neuroinflammation induced large oscillatory haemodynamic travelling waves in the frequency band of vasomotion (∼0.1 Hz) in both anaesthetized and awake rats. Vasomotor waves travelled through large distances between adjacent penetrating vessels, spanning the entire cortex thickness, and even extending to subcortical areas. Moreover, vasomotion amplitude correlated with microglial morphology changes and was significantly reduced by astrocytic toxins, suggesting that both microglia and astrocytes are involved in the enhancement of vasomotion during neuroinflammation. Notably, functional connectivity was increased under this oscillatory state and functional hyperaemia was exacerbated. INTERPRETATION These findings further reveal the spatiotemporal properties of cerebral vasomotion and suggest this is a major component of brain haemodynamics in pathological states. Moreover, reactive microglia and astrocytes are participating to increase vasomotion during neuroinflammation. For the field of functional neuroimaging, our results advocate for considering 0.1 Hz haemodynamic oscillations as an important complement to traditional measurements, particularly in neuroinflammatory conditions. Indeed, brain haemodynamics may provide insights not only into neuronal activity but also glial reactivity. FUNDING Supported by ANR ("LabCom-NI2D", "Labex Cortex") and Auvergne-Rhône-Alpes Region ("BI2D").
Collapse
Affiliation(s)
- Mickaël Pereira
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France
| | | | | | - Louis Vidal
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France
| | - Guillaume Marcy
- Université Claude Bernard Lyon 1, Bioinformatic Platform of the Labex Cortex, Lyon, France
| | - Sarah Benkeder
- Université Claude Bernard Lyon 1, Institut MeLiS, INSERM, CNRS, Lyon, France
| | - Paul Marchal
- Université Claude Bernard Lyon 1, Institut MeLiS, INSERM, CNRS, Lyon, France
| | - Jean-Christophe Comte
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France
| | - Olivier Pascual
- Université Claude Bernard Lyon 1, Institut MeLiS, INSERM, CNRS, Lyon, France
| | - Luc Zimmer
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France; CERMEP-Imaging Platform, Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Benjamin Vidal
- Université Claude Bernard Lyon 1, Lyon Neuroscience Research Center, INSERM, CNRS, Lyon, France; CERMEP-Imaging Platform, Lyon, France.
| |
Collapse
|
2
|
Li D, Huo X, Shen L, Qian M, Wang J, Mao S, Chen W, Li R, Zhu T, Zhang B, Liu K, Wu F, Bai Y. Astrocyte heterogeneity in ischemic stroke: Molecular mechanisms and therapeutic targets. Neurobiol Dis 2025; 209:106885. [PMID: 40139279 DOI: 10.1016/j.nbd.2025.106885] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Ischemic stroke is one of the major causes of death and disability in adults, bringing a significant economic burden to the society and families. Despite significant advancements in stroke treatment, focusing solely on neurons is insufficient for improving disease progression and prognosis. Astrocytes are the most ubiquitous cells in the brain, and they undergo morphological and functional changes after brain insults, which has been known as astrocyte reactivity. Transcriptomics have shown that reactive astrocytes (RA) are heterogeneous, and they can be roughly classified into neurotoxic and neuroprotective types, thereby affecting the development of central nervous system (CNS) diseases. However, the relationship between stroke and reactive astrocyte heterogeneity has not been fully elucidated, and regulating the heterogeneity of astrocytes to play a neuroprotective role may provide a new perspective for the treatment of stroke. Here we systematically review current advancements in astrocyte heterogeneity following ischemic stroke, elucidate the molecular mechanisms underlying their activation, and further summarize promising therapeutic agents and molecular targets capable of modulating astrocyte heterogeneity.
Collapse
Affiliation(s)
- Daxing Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xinchen Huo
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Shen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minjie Qian
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jindou Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shijie Mao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenjing Chen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Runheng Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Tianhao Zhu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Beicheng Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Kunxuan Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Feifei Wu
- Laboratory for Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
3
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
4
|
Poulen G, Douich N, Gazard CM, Mestre-Francés N, Cardoso M, Bauchet L, Vachiery-Lahaye F, Lonjon N, Gerber YN, Perrin FE. Sex and age differences in glia and myelin in nonhuman primate and human spinal cords: implications for pathology. Cell Death Discov 2025; 11:129. [PMID: 40175332 PMCID: PMC11965325 DOI: 10.1038/s41420-025-02425-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025] Open
Abstract
In a healthy central nervous system, glial cells are influenced by genetic, epigenetic, age, and sex factors. Aging typically causes astrocytes and microglia to undergo changes that reduce their neuroprotective functions and increase harmful activities. Additionally, sex-related differences in glial and myelin functions may impact neurological disorders. Despite this, few studies have investigated glial cells in primates, with most focusing on the brain. This study aims to explore whether glial cells and myelin exhibit age- and sex-related differences in the spinal cord of nonhuman primates and humans. We used immunohistochemistry and myelin staining to analyze healthy spinal cord samples from midlife and aged individuals of both sexes, focusing on Microcebus murinus (a small nonhuman primate) and humans. Primate spinal cords show distinct variations in glial markers and myelin characteristics related to sex and age, with differences varying between species. Notably, GFAP expression is sex-dependent in both primate species. We also observed greater differences in the expression of microglial markers than other glial markers. Overall, we found the opposite pattern for the g-ratio and oligodendrocytic marker between species. These findings suggest that glial cells may play a critical role in age- and sex-related differences in the prevalence and progression of spinal cord diseases.
Collapse
Affiliation(s)
- Gaëtan Poulen
- MMDN, Univ. Montpellier, EPHE, INSERM, Montpellier, France
- Department of Neurosurgery, CHU, Montpellier, France
| | - Nacéra Douich
- MMDN, Univ. Montpellier, EPHE, INSERM, Montpellier, France
| | - Chloé M Gazard
- MMDN, Univ. Montpellier, EPHE, INSERM, Montpellier, France
| | - Nadine Mestre-Francés
- MMDN, Univ. Montpellier, EPHE, INSERM, Montpellier, France
- PSL Research University, Paris, France
| | - Maïda Cardoso
- University of Montpellier, plateforme BNIF, Montpellier, France
| | - Luc Bauchet
- Department of Neurosurgery, CHU, Montpellier, France
- INSERM U1191, Institute of Functional Genomics, University of Montpellier, Montpellier, France
| | | | - Nicolas Lonjon
- MMDN, Univ. Montpellier, EPHE, INSERM, Montpellier, France
- Department of Neurosurgery, CHU, Montpellier, France
| | | | - Florence E Perrin
- MMDN, Univ. Montpellier, EPHE, INSERM, Montpellier, France.
- Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
5
|
Cha J, Zeng P, Zong H, Zhao J, Chen J, Zuo H, Zhang B, Shi C, Li J, Hua Q, Wang Z, Hou Y, Zhang R. Single-cell RNA sequencing of neonatal cortical astrocytes reveals versatile cell clusters during astrocyte-neuron conversion. Mol Biol Rep 2025; 52:189. [PMID: 39899158 DOI: 10.1007/s11033-025-10309-5] [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: 09/17/2024] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Astrocytes are extensively utilized as starting cells for neuronal conversion. Our previous study discovered that a portion of primary cultured mouse neonatal cortical astrocytes can be directly converted into neurons after exposure to a neurogenic induction condition. Recent in vivo studies have demonstrated astrocyte heterogeneity in terms of their developmental origin, molecular profile, physiology, and functional outputs. We hypothesized that the heterogeneity of primary astrocytes in our study could influence their conversion potential. METHODS AND RESULTS We performed single-cell RNA sequencing on cells harvested at key time points during in vitro astrocyte-to-neuron conversion, specifically on Day 1 and Day 9. Through single-cell RNA sequencing analysis, we identified several subpopulations of astrocytes, labeled as Astrocyte 1 to Astrocyte 3, based on distinct gene expression patterns. Pseudotime trajectory analysis predicted the existence of three distinct cell states throughout the conversion process. Astrocyte 3 exhibited a higher propensity for neuronal conversion, with proliferation genes like Mki67 being highly expressed. Additionally, several candidate genes were identified as potentially crucial in the conversion process. Astrocyte 3 is considered a unique subtype population of astrocytes. CONCLUSIONS Our investigation underscores the diversity of primary neonatal cortical astrocytes and provides critical insights into the potential for astrocyte-to-neuron conversion, which may be harnessed to enhance the efficiency of this astrocyte-neuron conversion process.
Collapse
Affiliation(s)
- Jiaxue Cha
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Peng Zeng
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hui Zong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiayi Zhao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiayao Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Haowei Zuo
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bowen Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Changjie Shi
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jing Li
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiuhong Hua
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zixin Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yujun Hou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Ru Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
6
|
Bao M, Wang X, Li X, Sun R, Wang Z, Jiang T, Wang H, Feng J. Single-Cell Landscape of the Cochlea Revealed Cell-Type-Specific Diversification in Hipposideros armiger Based on PacBio Long-Read Sequencing. Biomolecules 2025; 15:211. [PMID: 40001514 PMCID: PMC11853400 DOI: 10.3390/biom15020211] [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: 12/16/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Echolocation represents one of the most rapid adaptive sensorimotor modulation behaviors observed in mammals, establishing bats as one of the most evolutionarily successful mammals. Bats rely on high-frequency hearing for survival, but our understanding of its cellular molecular basis is scattered and segmented. Herein, we constructed the first single-cell transcriptomic landscape of the cochlea in Hipposideros armiger, a CF-FM bat, using a PacBio-optimized genome and compared it with the results obtained from unoptimized original genomes. Sixteen distinct cell types were distributed across five spatial regions of the cochlea. Notably, through hematoxylin and eosin staining and fluorescence in situ hybridization, we identified new types of spiral ganglion neuron (SGN) cells in the cochlea of H. armiger. These SGN cells are likely critical for auditory perception and may have driven the adaptive evolution of high-frequency hearing in this species. Furthermore, we uncovered the differentiation relationships of among specific cell types, such as the transition from supporting cells to hair cells. Using the cochlear cell atlas as a reference, cell types susceptible to deafness-associated genes (in the human) were also identified. In summary, this study provides novel insights into the cellular and molecular mechanisms underlying the adaptive high-frequency hearing in bats and highlights potential candidate cell types and genes for therapeutic interventions in hearing loss.
Collapse
Affiliation(s)
- Mingyue Bao
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xue Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xintong Li
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Ruyi Sun
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Zhiqiang Wang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Hui Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| |
Collapse
|
7
|
Sun J, Peng S, Yang Q, Yang J, Dai Y, Xing L. Microplastics/nanoplastics and neurological health: An overview of neurological defects and mechanisms. Toxicology 2025; 511:154030. [PMID: 39653181 DOI: 10.1016/j.tox.2024.154030] [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: 10/10/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
The widespread use of plastic products worldwide has brought about serious environmental issues. In natural environments, it's difficult for plastic products to degrade completely, and so they exist in the form of micro/nanoplastics (M/NPs), which have become a new type of pollutant. Prolonged exposure to M/NPs can lead to a series of health problems in humans, particularly toxicity to the nervous system, with consequences including neurodevelopmental abnormalities, neuronal death, neurological inflammation, and neurodegenerative diseases. Although direct evidence from humans is still limited, model organisms and organoids serve as powerful tools to provide important insights. This article summarizes the effects of M/NPs on the nervous system, focusing on cognitive function, neural development, and neuronal death. Mechanisms such as neurotransmitter synthesis and release, inflammatory responses, oxidative stress, the gut-brain axis, and the liver-brain axis are covered. The neurotoxicity induced by M/NPs may exacerbate or directly trigger neurodegenerative diseases and neurodevelopmental disorders. We particularly emphasize potential therapeutic agents that may counteract the neurotoxic effects induced by M/NPs, highlighting a novel future research direction. In summary, this paper cites evidence and provides mechanistic perspectives on the effects of M/NPs on neurological health, providing clues for eliminating M/NP hazards to human health in the future.
Collapse
Affiliation(s)
- Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University,Nantong, Jiangsu Province 226001, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University,Nantong, Jiangsu Province 226001, China
| | - Qiongxia Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University,Nantong, Jiangsu Province 226001, China
| | - Jiawei Yang
- Department of Neurology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu Province 226000, China
| | - Yanfei Dai
- Nantong Geriatric Rehabilitation Hospital, Branch of Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University,Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
8
|
Qiu J, Peng S, Qu R, Wu L, Xing L, Zhang L, Sun J. New evidence of vascular defects in neurodegenerative diseases revealed by single cell RNA sequencing. Clin Sci (Lond) 2024; 138:1377-1394. [PMID: 39469930 DOI: 10.1042/cs20241658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Neurodegenerative diseases (NDs) involve the progressive loss of neuronal structure or function in the brain and spinal cord. Despite their diverse etiologies, NDs manifest similar pathologies. Emerging research identifies vascular defects as a previously neglected hallmark of NDs. The development and popularization of single-cell RNA sequencing (scRNA-seq) technologies have significantly advanced our understanding of brain vascular cell types and their molecular characteristics, including gene expression changes at the single-cell level in NDs. These unprecedented insights deepen our understanding of the pathogenic mechanisms underlying NDs. However, the occurrence and role of vascular defects in disease progression remain largely unexplored. In this paper, we systematically summarize recent advances in the structure and organization of the central nervous system vasculature in mice, healthy individuals, and patients with NDs, focussing primarily on disease-specific alterations in vascular cell types or subtypes. Combining scRNA-seq with pathology evidence, we propose that vascular defects, characterized by disruptions in cell types and structural integrity, may serve as common early features of NDs. Finally, we discuss several pathways through which vascular defects in NDs lead to neuronal degeneration. A deeper understanding of the causes and contributions of vascular defects to NDs aids in elucidating the pathogenic mechanisms and developing meaningful therapeutic interventions.
Collapse
Affiliation(s)
- Jiaying Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
9
|
Wang J, Wang S, Li Q, Liu F, Wan Y, Liang H. Bibliometric and visual analysis of single-cell multiomics in neurodegenerative disease arrest studies. Front Neurol 2024; 15:1450663. [PMID: 39440247 PMCID: PMC11493674 DOI: 10.3389/fneur.2024.1450663] [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: 06/17/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Background Neurodegenerative diseases are progressive disorders that severely diminish the quality of life of patients. However, research on neurodegenerative diseases needs to be refined and deepened. Single-cell polyomics is a technique for obtaining transcriptomic, proteomic, and other information from a single cell. In recent years, the heat of single-cell multiomics as an emerging research tool for brain science has gradually increased. Therefore, the aim of this study was to analyze the current status and trends of studies related to the application of single-cell multiomics in neurodegenerative diseases through bibliometrics. Result A total of 596 publications were included in the bibliometric analysis. Between 2015 and 2022, the number of publications increased annually, with the total number of citations increasing significantly, exhibiting the fastest rate of growth between 2019 and 2022. The country/region collaboration map shows that the United States has the most publications and cumulative citations, and that China and the United States have the most collaborations. The institutions that produced the greatest number of articles were Harvard Medical School, Skupin, Alexander, and Wiendl. Among the authors, Heinz had the highest output. Mathys, H accumulated the most citations and was the authoritative author in the field. The journal Nature Communications has published the most literature in this field. A keyword analysis reveals that neurodegenerative diseases and lesions (e.g., Alzheimer's disease, amyloid beta) are the core and foundation of the field. Conversely, single-cell multiomics related research (e.g., single-cell RNA sequencing, bioinformatics) and brain nerve cells (e.g., microglia, astrocytes, neural stem cells) are the hot frontiers of this specialty. Among the references, the article "Single-cell transcriptomic analysis of Alzheimer's disease" is the most frequently cited (1,146 citations), and the article "Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq" was the most cited article in the field. Conclusion The objective of this study is to employ bibliometric methods to visualize studies related to single-cell multiomics in neurodegenerative diseases. This will enable us to summarize the current state of research and to reveal key trends and emerging hotspots in the field.
Collapse
Affiliation(s)
- Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, China
| | - Shuqing Wang
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Qingyu Li
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Fei Liu
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, China
| |
Collapse
|
10
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [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: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
11
|
Peng S, Cai X, Chen J, Sun J, Lai B, Chang M, Xing L. The role of CELF family in neurodevelopment and neurodevelopmental disorders. Neurobiol Dis 2024; 197:106525. [PMID: 38729272 DOI: 10.1016/j.nbd.2024.106525] [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: 02/10/2024] [Revised: 03/26/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024] Open
Abstract
RNA-binding proteins (RBPs) bind to RNAs and are crucial for regulating RNA splicing, stability, translation, and transport. Among these proteins, the CUGBP Elav-like family (CELF) is a highly conserved group crucial for posttranscriptional regulation by binding to CUG repeats. Comprising CELF1-6, this family exhibits diverse expression patterns and functions. Dysregulation of CELF has been implicated in various neural disorders, encompassing both neurodegenerative and neurodevelopmental conditions, such as Alzheimer's disease and autism. This article aims to provide a comprehensive summary of the CELF family's role in neurodevelopment and neurodevelopmental disorders. Understanding CELF's mechanisms may offer clues for potential therapeutic strategies by regulating their targets in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Xinyi Cai
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Junpeng Chen
- School of Nursing and Rehabilitation, Nantong University, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Biqin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Min Chang
- School of Education Science, Nantong University, Nantong 226019, China.
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China.
| |
Collapse
|
12
|
Nagayach A, Wang C. Autophagy in neural stem cells and glia for brain health and diseases. Neural Regen Res 2024; 19:729-736. [PMID: 37843206 PMCID: PMC10664120 DOI: 10.4103/1673-5374.382227] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 10/17/2023] Open
Abstract
Autophagy is a multifaceted cellular process that not only maintains the homeostatic and adaptive responses of the brain but is also dynamically involved in the regulation of neural cell generation, maturation, and survival. Autophagy facilities the utilization of energy and the microenvironment for developing neural stem cells. Autophagy arbitrates structural and functional remodeling during the cell differentiation process. Autophagy also plays an indispensable role in the maintenance of stemness and homeostasis in neural stem cells during essential brain physiology and also in the instigation and progression of diseases. Only recently, studies have begun to shed light on autophagy regulation in glia (microglia, astrocyte, and oligodendrocyte) in the brain. Glial cells have attained relatively less consideration despite their unquestioned influence on various aspects of neural development, synaptic function, brain metabolism, cellular debris clearing, and restoration of damaged or injured tissues. Thus, this review composes pertinent information regarding the involvement of autophagy in neural stem cells and glial regulation and the role of this connexion in normal brain functions, neurodevelopmental disorders, and neurodegenerative diseases. This review will provide insight into establishing a concrete strategic approach for investigating pathological mechanisms and developing therapies for brain diseases.
Collapse
Affiliation(s)
- Aarti Nagayach
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chenran Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
13
|
Liu J, Qi L, Bao S, Yan F, Chen J, Yu S, Dong C. The acute spinal cord injury microenvironment and its impact on the homing of mesenchymal stem cells. Exp Neurol 2024; 373:114682. [PMID: 38199509 DOI: 10.1016/j.expneurol.2024.114682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Spinal cord injury (SCI) is a highly debilitating condition that inflicts devastating harm on the lives of affected individuals, underscoring the urgent need for effective treatments. By activating inflammatory cells and releasing inflammatory factors, the secondary injury response creates an inflammatory microenvironment that ultimately determines whether neurons will undergo necrosis or regeneration. In recent years, mesenchymal stem cells (MSCs) have garnered increasing attention for their therapeutic potential in SCI. MSCs not only possess multipotent differentiation capabilities but also have homing abilities, making them valuable as carriers and mediators of therapeutic agents. The inflammatory microenvironment induced by SCI recruits MSCs to the site of injury through the release of various cytokines, chemokines, adhesion molecules, and enzymes. However, this mechanism has not been previously reported. Thus, a comprehensive exploration of the molecular mechanisms and cellular behaviors underlying the interplay between the inflammatory microenvironment and MSC homing is crucial. Such insights have the potential to provide a better understanding of how to harness the therapeutic potential of MSCs in treating inflammatory diseases and facilitating injury repair. This review aims to delve into the formation of the inflammatory microenvironment and how it influences the homing of MSCs.
Collapse
Affiliation(s)
- Jinyi Liu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Longju Qi
- Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Shengzhe Bao
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jiaxi Chen
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Shumin Yu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
14
|
Wu Z, Li G, Wang S, Zhang N, Li X, Zhang F, Niu J, Wang N, Zu J, Wang Y. Single-cell analysis of spinal cord injury reveals functional heterogeneity of oligodendrocyte lineage cells. Gene 2023; 886:147713. [PMID: 37579960 DOI: 10.1016/j.gene.2023.147713] [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: 06/17/2023] [Revised: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Spinal cord injury (SCI) is a traumatic condition that causes myelin destruction and neuronal death, making it challenging to reverse. In spinal cord tissue, oligodendrocyte progenitor cells and oligodendrocytes are essential for maintaining myelin morphology and axon regeneration. The decrease in oligodendrocyte lineage cells after SCI is a major factor contributing to the difficulty in restoring spinal cord function. However, there is still a lack of research on the status and intercellular communication between oligodendrocyte lineage cells after injury. The development of single-cell sequencing technology has enabled researchers to obtain highly accurate cellular transcriptional information, facilitating detailed studies of cellular subpopulations. This study delved into the cellular heterogeneity of oligodendrocyte lineage cells using a single-cell transcriptomic approach to uncover functional changes and cellular interactions during different time points after SCI. Our findings highlighted the critical roles of Psap (Prosaposin)/Gpr37l1 and Psap/Gpr37 ligand-receptor pairs among oligodendrocyte lineage cells. Furthermore, we predicted the transcription factors that may play a key regulatory role. We demonstrated for the first time that Junb acts almost exclusively in mature oligodendrocytes, which provides a potential target for the study of oligodendrocyte transcriptional mechanisms.
Collapse
Affiliation(s)
- Zexuan Wu
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guanglei Li
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Sikai Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Nan Zhang
- Department of Stomatology, Xiang An Hospital of Xiamen University, China
| | - Xuefeng Li
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Fawang Zhang
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiawen Niu
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Nanxiang Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jianing Zu
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Yufu Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
15
|
Lao Y, Li Z, Bai Y, Li W, Wang J, Wang Y, Li Q, Dong Z. Glial Cells of the Central Nervous System: A Potential Target in Chronic Prostatitis/Chronic Pelvic Pain Syndrome. Pain Res Manag 2023; 2023:2061632. [PMID: 38023826 PMCID: PMC10661872 DOI: 10.1155/2023/2061632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/24/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is one of the most common diseases of the male urological system while the etiology and treatment of CP/CPPS remain a thorny issue. Cumulative research suggested a potentially important role of glial cells in CP/CPPS. This narrative review retrospected literature and grasped the research process about glial cells and CP/CPPS. Three types of glial cells showed a crucial connection with general pain and psychosocial symptoms. Microglia might also be involved in lower urinary tract symptoms. Only microglia and astrocytes have been studied in the animal model of CP/CPPS. Activated microglia and reactive astrocytes were found to be involved in both pain and psychosocial symptoms of CP/CPPS. The possible mechanism might be to mediate the production of some inflammatory mediators and their interaction with neurons. Glial cells provide a new insight to understand the cause of complex symptoms of CP/CPPS and might become a novel target to develop new treatment options. However, the activation and action mechanism of glial cells in CP/CPPS needs to be further explored.
Collapse
Affiliation(s)
- Yongfeng Lao
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zewen Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanan Bai
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Weijia Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jian Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanan Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qingchao Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhilong Dong
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
16
|
Siddiq MM, Toro CA, Johnson NP, Hansen J, Xiong Y, Mellado W, Tolentino RE, Johnson K, Jayaraman G, Suhail Z, Harlow L, Dai J, Beaumont KG, Sebra R, Willis DE, Cardozo CP, Iyengar R. Spinal cord injury regulates circular RNA expression in axons. Front Mol Neurosci 2023; 16:1183315. [PMID: 37692100 PMCID: PMC10483835 DOI: 10.3389/fnmol.2023.1183315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/04/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Neurons transport mRNA and translational machinery to axons for local translation. After spinal cord injury (SCI), de novo translation is assumed to enable neurorepair. Knowledge of the identity of axonal mRNAs that participate in neurorepair after SCI is limited. We sought to identify and understand how axonal RNAs play a role in axonal regeneration. Methods We obtained preparations enriched in axonal mRNAs from control and SCI rats by digesting spinal cord tissue with cold-active protease (CAP). The digested samples were then centrifuged to obtain a supernatant that was used to identify mRNA expression. We identified differentially expressed genes (DEGS) after SCI and mapped them to various biological processes. We validated the DEGs by RT-qPCR and RNA-scope. Results The supernatant fraction was highly enriched for mRNA from axons. Using Gene Ontology, the second most significant pathway for all DEGs was axonogenesis. Among the DEGs was Rims2, which is predominately a circular RNA (circRNA) in the CNS. We show that Rims2 RNA within spinal cord axons is circular. We found an additional 200 putative circRNAs in the axonal-enriched fraction. Knockdown in primary rat cortical neurons of the RNA editing enzyme ADAR1, which inhibits formation of circRNAs, significantly increased axonal outgrowth and increased the expression of circRims2. Using Rims2 as a prototype we used Circular RNA Interactome to predict miRNAs that bind to circRims2 also bind to the 3'UTR of GAP-43, PTEN or CREB1, all known regulators of axonal outgrowth. Axonally-translated GAP-43 supports axonal elongation and we detect GAP-43 mRNA in the rat axons by RNAscope. Discussion By enriching for axonal RNA, we detect SCI induced DEGs, including circRNA such as Rims2. Ablation of ADAR1, the enzyme that regulates circRNA formation, promotes axonal outgrowth of cortical neurons. We developed a pathway model using Circular RNA Interactome that indicates that Rims2 through miRNAs can regulate the axonal translation GAP-43 to regulate axonal regeneration. We conclude that axonal regulatory pathways will play a role in neurorepair.
Collapse
Affiliation(s)
- Mustafa M. Siddiq
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Carlos A. Toro
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicholas P. Johnson
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jens Hansen
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yuguang Xiong
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Rosa E. Tolentino
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kaitlin Johnson
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
| | - Gomathi Jayaraman
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zaara Suhail
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lauren Harlow
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jinye Dai
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Robert Sebra
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dianna E. Willis
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Christopher P. Cardozo
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ravi Iyengar
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
17
|
Chu Y, Jia S, Xu K, Liu Q, Mai L, Liu J, Fan W, Huang F. Single-cell transcriptomic profile of satellite glial cells in trigeminal ganglion. Front Mol Neurosci 2023; 16:1117065. [PMID: 36818656 PMCID: PMC9932514 DOI: 10.3389/fnmol.2023.1117065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Satellite glial cells (SGCs) play an important role in regulating the function of trigeminal ganglion (TG) neurons. Multiple mediators are involved in the bidirectional communication between SGCs and neurons in different physiological and pathological states. However, molecular insights into the transcript characteristics of SGCs are limited. Moreover, little is known about the heterogeneity of SGCs in TG, and a more in-depth understanding of the interactions between SGCs and neuron subtypes is needed. Here we show the single-cell RNA sequencing (scRNA-seq) profile of SGCs in TG under physiological conditions. Our results demonstrate TG includes nine types of cell clusters, such as neurons, SGCs, myeloid Schwann cells (mSCs), non-myeloid Schwann cells (nmSCs), immune cells, etc., and the corresponding markers are also presented. We reveal the signature gene expression of SGCs, mSCs and nmSCs in the TG, and analyze the ligand-receptor pairs between neuron subtypes and SGCs in the TG. In the heterogeneity analysis of SGCs, four SGCs subtypes are identified, including subtypes enriched for genes associated with extracellular matrix organization, immediate early genes, interferon beta, and cell adhesion molecules, respectively. Our data suggest the molecular characteristics, heterogeneity of SGCs, and bidirectional interactions between SGCs and neurons, providing a valuable resource for studying SGCs in the TG.
Collapse
Affiliation(s)
- Yanhao Chu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shilin Jia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ke Xu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qing Liu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lijia Mai
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiawei Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China,*Correspondence: Wenguo Fan, ; Fang Huang,
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China,*Correspondence: Wenguo Fan, ; Fang Huang,
| |
Collapse
|
18
|
Gala DS, Titlow JS, Teodoro RO, Davis I. Far from home: the role of glial mRNA localization in synaptic plasticity. RNA (NEW YORK, N.Y.) 2023; 29:153-169. [PMID: 36442969 PMCID: PMC9891262 DOI: 10.1261/rna.079422.122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Neurons and glia are highly polarized cells, whose distal cytoplasmic functional subdomains require specific proteins. Neurons have axonal and dendritic cytoplasmic extensions containing synapses whose plasticity is regulated efficiently by mRNA transport and localized translation. The principles behind these mechanisms are equally attractive for explaining rapid local regulation of distal glial cytoplasmic projections, independent of their cell nucleus. However, in contrast to neurons, mRNA localization has received little experimental attention in glia. Nevertheless, there are many functionally diverse glial subtypes containing extensive networks of long cytoplasmic projections with likely localized regulation that influence neurons and their synapses. Moreover, glia have many other neuron-like properties, including electrical activity, secretion of gliotransmitters and calcium signaling, influencing, for example, synaptic transmission, plasticity and axon pruning. Here, we review previous studies concerning glial transcripts with important roles in influencing synaptic plasticity, focusing on a few cases involving localized translation. We discuss a variety of important questions about mRNA transport and localized translation in glia that remain to be addressed, using cutting-edge tools already available for neurons.
Collapse
Affiliation(s)
- Dalia S Gala
- Department of Biochemistry, The University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Joshua S Titlow
- Department of Biochemistry, The University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Rita O Teodoro
- iNOVA4Health, NOVA Medical School-Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa 1169-056, Portugal
| | - Ilan Davis
- Department of Biochemistry, The University of Oxford, Oxford OX1 3QU, United Kingdom
| |
Collapse
|
19
|
Early Diagnosis of Brain Diseases Using Artificial Intelligence and EV Molecular Data: A Proposed Noninvasive Repeated Diagnosis Approach. Cells 2022; 12:cells12010102. [PMID: 36611896 PMCID: PMC9818301 DOI: 10.3390/cells12010102] [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: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Brain-derived extracellular vesicles (BDEVs) are released from the central nervous system. Brain-related research and diagnostic techniques involving BDEVs have rapidly emerged as a means of diagnosing brain disorders because they are minimally invasive and enable repeatable measurements based on body fluids. However, EVs from various cells and organs are mixed in the blood, acting as potential obstacles for brain diagnostic systems using BDEVs. Therefore, it is important to screen appropriate brain EV markers to isolate BDEVs in blood. Here, we established a strategy for screening potential BDEV biomarkers. To collect various molecular data from the BDEVs, we propose that the sensitivity and specificity of the diagnostic system could be enhanced using machine learning and AI analysis. This BDEV-based diagnostic strategy could be used to diagnose various brain diseases and will help prevent disease through early diagnosis and early treatment.
Collapse
|
20
|
Cao Y, Zhu S, Yu B, Yao C. Single-cell RNA sequencing for traumatic spinal cord injury. FASEB J 2022; 36:e22656. [PMID: 36374259 DOI: 10.1096/fj.202200943r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/28/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Traumatic spinal cord injury (tSCI) is a severe injury of the central nervous system (CNS) with complicated pathological microenvironment that results in hemorrhage, inflammation, and scar formation. The microenvironment of the injured spinal cord comprises heterogeneous neurons, glial cells, inflammatory cells, and stroma-related cells. Increasing evidence has indicated that the altered cellular and molecular microenvironment following tSCI is a key factor impeding functional recovery. Single-cell RNA sequencing (scRNA-seq) has provided deep insights into the dynamic cellular and molecular changes in the microenvironment by comprehensively characterizing the diversity of spinal cord cell types. Specifically, scRNA-seq enables the exploration of the molecular mechanisms underlying tSCI by elucidating intercellular communication in spinal cord samples between normal and injury conditions at a single-cell resolution. Here, we first described the pathological and physiological processes after tSCI and gave a brief introduction of the scRNA-seq technology. We then focused on the recent scRNA-seq researches in tSCI, which characterized diverse cell-type populations and specific cell-cell interactions in tSCI. In addition, we also highlighted some potential directions for the research of scRNA-seq in tSCI in the future.
Collapse
Affiliation(s)
- Yuqi Cao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shunxing Zhu
- Laboratory Animals Center, Nantong University, Nantong, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
21
|
Zhao D, Zhang Y, Zheng Y, Li XT, Sun CC, Yang Q, Xie Q, Xu DS. Double-target neural circuit-magnetic stimulation improves motor function in spinal cord injury by attenuating astrocyte activation. Neural Regen Res 2022; 18:1062-1066. [PMID: 36254994 PMCID: PMC9827772 DOI: 10.4103/1673-5374.355768] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Multi-target neural circuit-magnetic stimulation has been clinically shown to improve rehabilitation of lower limb motor function after spinal cord injury. However, the precise underlying mechanism remains unclear. In this study, we performed double-target neural circuit-magnetic stimulation on the left motor cortex and bilateral L5 nerve root for 3 successive weeks in a rat model of incomplete spinal cord injury caused by compression at T10. Results showed that in the injured spinal cord, the expression of the astrocyte marker glial fibrillary acidic protein and inflammatory factors interleukin 1β, interleukin-6, and tumor necrosis factor-α had decreased, whereas that of neuronal survival marker microtubule-associated protein 2 and synaptic plasticity markers postsynaptic densification protein 95 and synaptophysin protein had increased. Additionally, neural signaling of the descending corticospinal tract was markedly improved and rat locomotor function recovered significantly. These findings suggest that double-target neural circuit-magnetic stimulation improves rat motor function by attenuating astrocyte activation, thus providing a theoretical basis for application of double-target neural circuit-magnetic stimulation in the clinical treatment of spinal cord injury.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Rehabilitation, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ye Zhang
- Department of Rehabilitation, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ya Zheng
- Department of Rehabilitation, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xu-Tong Li
- Department of Neurology, Zibo Centre Hospital, Zibo, Shandong Province, China
| | - Cheng-Cheng Sun
- Department of Rehabilitation, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qi Yang
- Department of Rehabilitation, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qing Xie
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Correspondence to: Qing Xie, ; Dong-Sheng Xu, .
| | - Dong-Sheng Xu
- Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Shanghai, China,Correspondence to: Qing Xie, ; Dong-Sheng Xu, .
| |
Collapse
|
22
|
Qiu J, Wu L, Qu R, Jiang T, Bai J, Sheng L, Feng P, Sun J. History of development of the life-saving drug "Nusinersen" in spinal muscular atrophy. Front Cell Neurosci 2022; 16:942976. [PMID: 36035257 PMCID: PMC9414009 DOI: 10.3389/fncel.2022.942976] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disorder with an incidence of 1/6,000-1/10,000 and is the leading fatal disease among infants. Previously, there was no effective treatment for SMA. The first effective drug, nusinersen, was approved by the US FDA in December 2016, providing hope to SMA patients worldwide. The drug was introduced in the European Union in 2017 and China in 2019 and has so far saved the lives of several patients in most parts of the world. Nusinersen are fixed sequence antisense oligonucleotides with special chemical modifications. The development of nusinersen progressed through major scientific discoveries in medicine, genetics, biology, and other disciplines, wherein several scientists have made substantial contributions. In this article, we will briefly describe the pathogenesis and therapeutic strategies of SMA, summarize the timeline of important scientific findings during the development of nusinersen in a detailed, scientific, and objective manner, and finally discuss the implications of the development of nusinersen for SMA research.
Collapse
Affiliation(s)
- Jiaying Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong, China
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, China
| | - Tao Jiang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jialin Bai
- College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Pengchao Feng
- Nanjing Antisense Biopharmaceutical Co., Ltd, Nanjing, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| |
Collapse
|
23
|
Feng Y, Peng Y, Jie J, Yang Y, Yang P. The immune microenvironment and tissue engineering strategies for spinal cord regeneration. Front Cell Neurosci 2022; 16:969002. [PMID: 35990891 PMCID: PMC9385973 DOI: 10.3389/fncel.2022.969002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regeneration of neural tissue is limited following spinal cord injury (SCI). Successful regeneration of injured nerves requires the intrinsic regenerative capability of the neurons and a suitable microenvironment. However, the local microenvironment is damaged, including insufficient intraneural vascularization, prolonged immune responses, overactive immune responses, dysregulated bioenergetic metabolism and terminated bioelectrical conduction. Among them, the immune microenvironment formed by immune cells and cytokines plays a dual role in inflammation and regeneration. Few studies have focused on the role of the immune microenvironment in spinal cord regeneration. Here, we summarize those findings involving various immune cells (neutrophils, monocytes, microglia and T lymphocytes) after SCI. The pathological changes that occur in the local microenvironment and the function of immune cells are described. We also summarize and discuss the current strategies for treating SCI with tissue-engineered biomaterials from the perspective of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Feng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yong Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jing Jie
- Department of Clinical Laboratory, The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong, China
- Jing Jie,
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Yumin Yang,
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin Medical University, Harbin, China
- *Correspondence: Pengxiang Yang,
| |
Collapse
|
24
|
Kimball EC, Quillen S, Pease ME, Keuthan C, Nagalingam A, Zack DJ, Johnson TV, Quigley HA. Aquaporin 4 is not present in normal porcine and human lamina cribrosa. PLoS One 2022; 17:e0268541. [PMID: 35709078 PMCID: PMC9202842 DOI: 10.1371/journal.pone.0268541] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/03/2022] [Indexed: 02/02/2023] Open
Abstract
Aquaporin 4 is absent from astrocytes in the rodent optic nerve head, despite high expression in the retina and myelinated optic nerve. The purpose of this study was to quantify regional aquaporin channel expression in astrocytes of the porcine and human mouse optic nerve (ON). Ocular tissue sections were immunolabeled for aquaporins 1(AQP1), 4(AQP4), and 9(AQP9), myelin basic protein (MBP), glial fibrillary acidic protein (GFAP) and alpha-dystroglycan (αDG) for their presence in retina, lamina, myelin transition zone (MTZ, region just posterior to lamina) and myelinated ON (MON). Semi- quantification of AQP4 labeling & real-time quantitative PCR (qPCR) data were analyzed in retina and ON tissue. Porcine and control human eyes had abundant AQP4 in Müller cells, retinal astrocytes, and myelinated ON (MON), but minimal expression in the lamina cribrosa. AQP1 and AQP9 were present in retina, but not in the lamina. Immunolabeling of GFAP and αDG was similar in lamina, myelin transition zone (MTZ) and MON regions. Semi-quantitative AQP4 labeling was at background level in lamina, increasing in the MTZ, and highest in the MON (lamina vs MTZ, MON; p≤0.05, p≤0.01, respectively). Expression of AQP4 mRNA was minimal in lamina and substantial in MTZ and MON, while GFAP mRNA expression was uniform among the lamina, MTZ, and MON regions. Western blot assay showed AQP4 protein expression in the MON samples, but none was detected in the lamina tissue. The minimal presence of AQP4 in the lamina is a specific regional phenotype of astrocytes in the mammalian optic nerve head.
Collapse
Affiliation(s)
- Elizabeth C. Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sarah Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mary E. Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Casey Keuthan
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Aru Nagalingam
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Donald J. Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Thomas V. Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Harry A. Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
25
|
Sun J, Ji Y, Liang Q, Ming M, Chen Y, Zhang Q, Zhou S, Shen M, Ding F. Expression of Protein Acetylation Regulators During Peripheral Nerve Development, Injury, and Regeneration. Front Mol Neurosci 2022; 15:888523. [PMID: 35663264 PMCID: PMC9157241 DOI: 10.3389/fnmol.2022.888523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Protein acetylation, regulated by acetyltransferases and deacetylases, is an important post-translational modification that is involved in numerous physiological and pathological changes in peripheral nerves. There is still no systematical analysis on the expression changes of protein acetylation regulators during sciatic nerve development, injury, and regeneration. Here, we sequenced and analyzed the transcriptome of mouse sciatic nerves during development and after injury. We found that the changes in the expression of most regulators followed the rule that “development is consistent with regeneration and opposite to injury.” Immunoblotting with pan-acetylated antibodies also revealed that development and regeneration are a process of increased acetylation, while injury is a process of decreased acetylation. Moreover, we used bioinformatics methods to analyze the possible downstream molecules of two key regulators, histone deacetylase 1 (Hdac1) and lysine acetyltransferase 2b (Kat2b), and found that they were associated with many genes that regulate the cell cycle. Our findings provide an insight into the association of sciatic nerve development, injury, and regeneration from the perspective of protein acetylation.
Collapse
|