1
|
Wang Q, Zeng F, Fang C, Sun Y, Zhao X, Rong X, Zhang H, Xu Y. Galectin-3 alleviates demyelination by modulating microglial anti-inflammatory polarization through PPARγ-CD36 axis. Brain Res 2024; 1842:149106. [PMID: 38986827 DOI: 10.1016/j.brainres.2024.149106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
Demyelination is characterized by disruption of myelin sheath and disorders in myelin formation. Currently, there are no effective therapeutic treatments available. Microglia, especially anti-inflammatory phenotype microglia are critical for remyelination. Galectin-3 (Gal-3), which is known to modulate microglia activation, is correlated with myelination. In this study, we aimed to elucidate the roles of Gal-3 during myelin formation and explore the efficiency and mechanism of rGal-3 administration in remyelination. We enrolled Gal-3 knockout (Lgals3 KO) mice and demonstrated Lgals3 KO causes demyelination during spontaneous myelinogenesis. We performed a cuprizone (CPZ) intoxication model and found Lgals3 KO aggravates demyelinated lesions and favors microglial pro-inflammatory phenotype polarization. Recombinant Gal-3 (rGal-3) administration alleviates CPZ intoxication and drives microglial towards anti-inflammatory phenotype. Additionally, RNA sequencing results reveal the correlation between Gal-3 and the PPARγ-CD36 axis. Thus, we performed SSO and GW9662 administration to inhibit the activation of the PPARγ-CD36 axis and found that rGal-3 administration modulates microglial phenotype polarization by regulating the PPARγ-CD36 axis. Together, our findings highlight the importance of Gal-3 in myelination and provide insights into rGal-3 administration for modulating microglial anti-inflammatory phenotype polarization through the PPARγ-CD36 axis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China; Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China.
| | - Fansen Zeng
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Chunxiao Fang
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Yi Sun
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Xiaopeng Zhao
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Xiao Rong
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Huayan Zhang
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China.
| | - Yi Xu
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China.
| |
Collapse
|
2
|
Xing YL, Grossauer S, Park JW, Nasajpour E, Bui B, Morales D, Panovska D, Nirschl JJ, Feng ZP, Wei R, Koeck K, Thomason W, Xiu J, Harter PN, Filipski K, Mahaney K, Ji X, Mulcahy Levy JM, Grant GA, Prolo LM, Walsh KM, Lim M, Hambardzumyan D, Petritsch CK. Dual MAPK Inhibition Triggers Pro-inflammatory Signals and Sensitizes BRAF V600E Glioma to T Cell-Mediated Checkpoint Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.03.526065. [PMID: 39416185 PMCID: PMC11482820 DOI: 10.1101/2023.02.03.526065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
BRAF V600E pediatric low-grade gliomas frequently transform into high-grade gliomas (HGG) and poorly respond to chemotherapy, resulting in high mortality. Although combined BRAF and MEK inhibition (BRAFi+MEKi) outperforms chemotherapy, ∼70% of BRAF V600E HGG patients are therapy resistant and undergo unbridled tumor progression. BRAF V600E glioma have an immune-rich microenvironment suggesting that they could be responsive to immunotherapy but effects of BRAFi+MEKi on anti-tumor immunity are unclear. Using patient tumor tissue before and after BRAFi+MEKi, two novel syngeneic murine models of BRAF V600E HGG, and patient-derived cell lines, we examined the effects of clinically relevant BRAFi+MEKi with dabrafenib and trametinib on tumor growth, cell states, and tumor-infiltrating T cells. We find that BRAFi+MEKi treatment: i) upregulated programmed cell death protein-1 (PD-1) signaling genes and PD-1 ligand (PD-L1) protein expression in murine BRAF V600E HGG by stimulating IFNγ and IL-27, ii) attenuated T cell activity by IL-23, IL-27 and IL-32 production, which can promote the expansion of regulatory T cells, and iii) induced glial differentiation linked to a therapy-resistant PD-L1+ compartment through Galectin-3 secretion by tumor cells. Murine BRAF V600E HGG shrinkage by BRAFi+MEKi is associated with the upregulation of interferon-gamma response genes, MHC class I/II expression, and antigen presentation and processing programs, indicative of increased anti-tumor immunity. Combined BRAFi+MEKi with therapeutic antibodies inhibiting the PD-1 and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) immune checkpoints re-activate T cells and provide a survival benefit over single therapy in a T cell-dependent manner. The quadruple treatment overcame BRAFi+MEKi resistance by invigorating T cell-mediated anti-tumor immunity in murine BRAF V600E HGG. PD-L1 expression was elevated in human BRAF-mutant versus BRAF-wildtype glioblastoma clinical specimen, complementing experimental findings and suggesting translational relevance for patient care.
Collapse
|
3
|
Mayo F, González-Vinceiro L, Hiraldo-González L, Calle-Castillejo C, Torres-Rubio I, Mayo M, Ramírez-Lorca R, Echevarría M. Absence of Aquaporin-4 (AQP4) Prolongs the Presence of a CD11c+ Microglial Population during Postnatal Corpus Callosum Development. Int J Mol Sci 2024; 25:8332. [PMID: 39125902 PMCID: PMC11312288 DOI: 10.3390/ijms25158332] [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: 06/29/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Aquaporin-4 (AQP4) expression is associated with the development of congenital hydrocephalus due to its structural role in the ependymal membrane. Gene expression analysis of periaqueductal tissue in AQP4-knockout (KO) mice at 11 days of age (P11) showed a modification in ependymal cell adhesion and ciliary protein expression that could alter cerebrospinal fluid homeostasis. A microglial subpopulation of CD11c+ cells was overexpressed in the periaqueductal tissue of mice that did not develop hydrocephalus, suggesting a possible protective effect. Here, we verified the location of this CD11c+ expression in the corpus callosum (CC) and cerebellum of AQP4-KO mice and analysed its time course. Immunofluorescence labelling of the CD11c protein in the CC and cerebellum of WT and KO animals at P3, P5, P7 and P11 confirmed an expanded presence of these cells in both tissues of the KO animal; CD11c+ cells appeared at P3 and reached a peak at P11, whereas in the WT animal, they appeared at P5, reached their peak at P7 and were undetectable by P11. The gene expression analysis in the CC samples at P11 confirmed the presence of CD11c+ microglial cells in this tissue. Among the more than 4000 overexpressed genes, Spp1 stood out with the highest differential gene expression (≅600), with other genes, such as Gpnmb, Itgax, Cd68 and Atp6v0d2, also identified as overexpressed. Therefore, CD11c+ cells appear to be necessary for normal corpus callosum development during postnatal life, and the absence of AQP4 prolonged its expression in this tissue.
Collapse
Affiliation(s)
- Francisco Mayo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Lourdes González-Vinceiro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Laura Hiraldo-González
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Claudia Calle-Castillejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Ismael Torres-Rubio
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Manuel Mayo
- Física Teórica, Universidad de Sevilla, Apartado de Correos 1065, 41080 Sevilla, Spain
| | - Reposo Ramírez-Lorca
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Miriam Echevarría
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| |
Collapse
|
4
|
Funaki M, Nio-Kobayashi J, Suzuki R, Bando Y. Galectin-3 Plays a Role in Neuroinflammation in the Visual Pathway in Experimental Optic Neuritis. Cells 2024; 13:612. [PMID: 38607051 PMCID: PMC11011492 DOI: 10.3390/cells13070612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 04/13/2024] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) featuring numerous neuropathologies, including optic neuritis (ON) in some patients. However, the molecular mechanisms of ON remain unknown. Galectins, β-galactoside-binding lectins, are involved in various pathophysiological processes. We previously showed that galectin-3 (gal-3) is associated with the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. In the current study, we investigated the expression of gal-3 in the visual pathway in EAE mice to clarify its role in the pathogenesis of ON. Immunohistochemical analysis revealed upregulation of gal-3 in the visual pathway of the EAE mice during the peak stage of the disease, compared with naïve and EAE mice during the chronic stage. Gal-3 was detected mainly in microglia/macrophages and astrocytes in the visual pathway in EAE mice. In addition, gal-3+/Iba-1+ cells, identified as phagocytic by immunostaining for cathepsin D, accumulated in demyelinating lesions in the visual pathway during the peak disease stage of EAE. Moreover, NLRP3 expression was detected in most gal-3+/Iba-1+ cells. These results strongly suggest that gal-3 regulates NLRP3 signaling in microglia/macrophages and neuroinflammatory demyelination in ON. In astrocytes, gal-3 was expressed from the peak to the chronic disease stages. Taken together, our findings suggest a critical role of gal-3 in the pathogenesis of ON. Thus, gal-3 in glial cells may serve as a potential therapeutic target for ON.
Collapse
Affiliation(s)
- Masako Funaki
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Junko Nio-Kobayashi
- Department of Functional Glycobiology in Infectious Diseases, National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki 852-8523, Japan
| | - Ryoji Suzuki
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Yoshio Bando
- Department of Anatomy, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| |
Collapse
|
5
|
Carver CM, Gomez PT, Rodriguez SL, Kachergus JM, Liu Y, Shi J, Tran T, Wang L, Melov S, Thompson EA, Schafer MJ. Senescent and disease-associated microglia are modifiable features of aged brain white matter. RESEARCH SQUARE 2023:rs.3.rs-3467812. [PMID: 37961365 PMCID: PMC10635389 DOI: 10.21203/rs.3.rs-3467812/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Brain white matter tracts undergo structural and functional changes linked to late-life cognitive decline, but the cellular and molecular contributions to their selective vulnerability are not well defined. In naturally aged mice, we demonstrate that senescent and disease-associated microglia (DAM) phenotypes converge in hippocampus-adjacent white matter. Through gold-standard gene expression and immunolabeling combined with high-dimensional spatial mapping, we identified microglial cell fates in aged white matter characterized by aberrant morphology, microenvironment reorganization, and expression of senescence and DAM markers, including galectin 3 (GAL3/Lgals3), B-cell lymphoma 2 (Bcl2), and cyclin dependent kinase inhibitors, including Cdkn2a/p16ink4a. Pharmacogenetic or pharmacological targeting of p16ink4a or BCL2 reduced white matter GAL3+ DAM abundance and rejuvenated microglial fimbria organization. Our results demonstrate dynamic changes in microglial identity in aged white matter that can be reverted by senotherapeutic intervention to promote homeostatic maintenance in the aged brain.
Collapse
Affiliation(s)
- Chase M. Carver
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Paul T. Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Sonia L. Rodriguez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | | | - Yi Liu
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Ji Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Tommy Tran
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Marissa J. Schafer
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
6
|
Zhou Z, Feng Z, Sun X, Wang Y, Dou G. The Role of Galectin-3 in Retinal Degeneration and Other Ocular Diseases: A Potential Novel Biomarker and Therapeutic Target. Int J Mol Sci 2023; 24:15516. [PMID: 37958500 PMCID: PMC10649114 DOI: 10.3390/ijms242115516] [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: 10/09/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Galectin-3 is the most studied member of the Galectin family, with a large range of mediation in biological activities such as cell growth, proliferation, apoptosis, differentiation, cell adhesion, and tissue repair, as well as in pathological processes such as inflammation, tissue fibrosis, and angiogenesis. As is known to all, inflammation, aberrant cell apoptosis, and neovascularization are the main pathophysiological processes in retinal degeneration and many ocular diseases. Therefore, the review aims to conclude the role of Gal3 in the retinal degeneration of various diseases as well as the occurrence and development of the diseases and discuss its molecular mechanisms according to research in systemic diseases. At the same time, we summarized the predictive role of Gal3 as a biomarker and the clinical application of its inhibitors to discuss the possibility of Gal3 as a novel target for the treatment of ocular diseases.
Collapse
Affiliation(s)
| | | | | | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| | - Guorui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| |
Collapse
|
7
|
Patil V, Bohara R, Krishna Kanala V, McMahon S, Pandit A. Models and approaches to comprehend and address glial inflammation following spinal cord injury. Drug Discov Today 2023; 28:103722. [PMID: 37482236 DOI: 10.1016/j.drudis.2023.103722] [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: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Spinal cord injury (SCI) culminates in chronic inflammation and glial scar formation driven by the activation of microglia and astrocytes. Current anti-inflammatory strategies to treat glial activation associated with SCI have several limitations. Existing in vitro and ex vivo models studying molecular mechanisms associated with inflammation focus only on the acute phase. However, the progression of glial cell-derived inflammation over the acute-to-chronic phases has not been assessed. Understanding this progression will help establish a framework for evaluating therapeutic strategies. Additionally, new models could be useful as high-throughput screening (HTS) platforms. This review aims to highlight currently available models and future methods that could facilitate screening of novel therapeutics for SCI.
Collapse
Affiliation(s)
- Vaibhav Patil
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Vijaya Krishna Kanala
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Siobhan McMahon
- Anatomy, School of Medicine, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
8
|
Luo M, Qiu Z, Tang X, Wu L, Li S, Zhu J, Jiang Y. Inhibiting Cyclin B1-treated Pontine Infarction by Suppressing Proliferation of SPP1+ Microglia. Mol Neurobiol 2023; 60:1782-1796. [PMID: 36572839 DOI: 10.1007/s12035-022-03183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022]
Abstract
Pontine infarction is the major subtype of brainstem stroke causing severe neurological deficits. The pathophysiology and treatment of pontine infarction was rarely studied. A rat model of acute pontine infarction was established via injection of endothelin-1 in the pons. Single-cell RNA sequencing was applied to detect the cellular response in pontine infarction. Based on this finding, a potential treatment for pontine infarction targeting microglia was verified. Occlusion of penetrating artery caused by endothelin-1 led to pontine infarction. Single-cell RNA sequencing revealed a subtype of activated microglia, SPP1+ microglia, which were different from M1-like or M2-like depolarization. SPP1+ microglia interacted with oligodendrocytes and contributed to the demyelination of nerve tracts. Cyclin B1 regulated the proliferation of SPP1+ microglia. Cucurbitacin E, a cyclin B1 inhibitor, reduced the proliferation of SPP1+ microglia around the injured myelin sheath and alleviated the demyelination. Moreover, cucurbitacin E treatment decreased the ischemic infarction volume and neurological deficits after pontine infarction. SPP1+ microglia contributed to axonal demyelination in the pontine infarction, and inhibition of SPP1+ microglia provided neuroprotection for pontine infarction.
Collapse
Affiliation(s)
- Ming Luo
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China
| | - Zhihua Qiu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China
| | - Xiangyue Tang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China
| | - Li Wu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China
| | - Shaojun Li
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, 215300, China.
| | - Yongjun Jiang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China.
| |
Collapse
|
9
|
Xu J, Zhang L, Li M, He X, Luo J, Wu R, Hong Z, Zheng H, Hu X. TREM2 mediates physical exercise-promoted neural functional recovery in rats with ischemic stroke via microglia-promoted white matter repair. J Neuroinflammation 2023; 20:50. [PMID: 36829205 PMCID: PMC9960657 DOI: 10.1186/s12974-023-02741-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/17/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND The repair of white matter injury is of significant importance for functional recovery after ischemic stroke, and the up-regulation of triggering receptors expressed on myeloid cells 2 (TREM2) after ischemic stroke is neuroprotective and implicated in remyelination. However, the lack of effective therapies calls for the need to investigate the regenerative process of remyelination and the role of rehabilitation therapy. This study sought to investigate whether and how moderate physical exercise (PE) promotes oligodendrogenesis and remyelination in rats with transient middle cerebral artery occlusion (tMCAO). METHODS Male Sprague-Dawley rats (weighing 250-280 g) were subjected to tMCAO. AAV-shRNA was injected into the lateral ventricle to silence the Trem2 gene before the operation. The rats in the physical exercise group started electric running cage training at 48 h after the operation. The Morris water maze and novel object recognition test were used to evaluate cognitive function. Luxol fast blue staining, diffusion tensor imaging, and electron microscopy were used to observe myelin injury and repair. Immunofluorescence staining was applied to observe the proliferation and differentiation of oligodendrocyte precursor cells (OPCs). Expression of key molecules were detected using immunofluorescence staining, quantitative real-time polymerase chain reaction, Western blotting, and Enzyme-linked immunosorbent assay, respectively. RESULTS PE exerted neuroprotective efects by modulating microglial state, promoting remyelination and recovery of neurological function of rats over 35 d after stroke, while silencing Trem2 expression in rats suppressed the aforementioned effects promoted by PE. In addition, by leveraging the activin-A neutralizing antibody, we found a direct beneficial effect of PE on microglia-derived activin-A and its subsequent role on oligodendrocyte differentiation and remyelination mediated by the activin-A/Acvr axis. CONCLUSIONS The present study reveals a novel regenerative role of PE in white matter injury after stroke, which is mediated by upregulation of TREM2 and microglia-derived factor for oligodendrocytes regeneration. PE is an effective therapeutic approach for improving white matter integrity and alleviating neurological function deficits after ischemic stroke.
Collapse
Affiliation(s)
- Jinghui Xu
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Liying Zhang
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Mingyue Li
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Xiaofei He
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Jing Luo
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Rui Wu
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Zhongqiu Hong
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| |
Collapse
|
10
|
Canto-Gomes J, Da Silva-Ferreira S, Silva CS, Boleixa D, Martins da Silva A, González-Suárez I, Cerqueira JJ, Correia-Neves M, Nobrega C. People with Primary Progressive Multiple Sclerosis Have a Lower Number of Central Memory T Cells and HLA-DR + Tregs. Cells 2023; 12:439. [PMID: 36766781 PMCID: PMC9913799 DOI: 10.3390/cells12030439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
The importance of circulating immune cells to primary progressive multiple sclerosis (PPMS) pathophysiology is still controversial because most immunotherapies were shown to be ineffective in treating people with PPMS (pwPPMS). Yet, although controversial, data exist describing peripheral immune system alterations in pwPPMS. This study aims to investigate which alterations might be present in pwPPMS free of disease-modifying drugs (DMD) in comparison to age- and sex-matched healthy controls. A multicentric cross-sectional study was performed using 23 pwPPMS and 23 healthy controls. The phenotype of conventional CD4+ and CD8+ T cells, regulatory T cells (Tregs), B cells, natural killer (NK) T cells and NK cells was assessed. Lower numbers of central memory CD4+ and CD8+ T cells and activated HLA-DR+ Tregs were observed in pwPPMS. Regarding NK and NKT cells, pwPPMS presented higher percentages of CD56dimCD57+ NK cells expressing NKp46 and of NKT cells expressing KIR2DL2/3 and NKp30. Higher disease severity scores and an increasing time since diagnosis was correlated with lower numbers of inhibitory NK cells subsets. Our findings contribute to reinforcing the hypotheses that alterations in peripheral immune cells are present in pwPPMS and that changes in NK cell populations are the strongest correlate of disease severity.
Collapse
Affiliation(s)
- João Canto-Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Sara Da Silva-Ferreira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Carolina S. Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Division of Infectious Diseases, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden
| | | | - Ana Martins da Silva
- Porto University Hospital Center, 4099-001 Porto, Portugal
- Multidisciplinary Unit for Biomedical Research (UMIB)—ICBAS, University of Porto, 4050-346 Porto, Portugal
| | - Inés González-Suárez
- University Hospital Complex of Vigo, 36312 Vigo, Spain
- Álvaro Cunqueiro Hospital, 36312 Vigo, Spain
| | - João J. Cerqueira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Hospital of Braga, 4710-243 Braga, Portugal
- Clinical Academic Centre, Hospital of Braga, 4710-243 Braga, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Division of Infectious Diseases, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Claudia Nobrega
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
11
|
Raas Q, Tawbeh A, Tahri-Joutey M, Gondcaille C, Keime C, Kaiser R, Trompier D, Nasser B, Leoni V, Bellanger E, Boussand M, Hamon Y, Benani A, Di Cara F, Truntzer C, Cherkaoui-Malki M, Andreoletti P, Savary S. Peroxisomal defects in microglial cells induce a disease-associated microglial signature. Front Mol Neurosci 2023; 16:1170313. [PMID: 37138705 PMCID: PMC10149961 DOI: 10.3389/fnmol.2023.1170313] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Microglial cells ensure essential roles in brain homeostasis. In pathological condition, microglia adopt a common signature, called disease-associated microglial (DAM) signature, characterized by the loss of homeostatic genes and the induction of disease-associated genes. In X-linked adrenoleukodystrophy (X-ALD), the most common peroxisomal disease, microglial defect has been shown to precede myelin degradation and may actively contribute to the neurodegenerative process. We previously established BV-2 microglial cell models bearing mutations in peroxisomal genes that recapitulate some of the hallmarks of the peroxisomal β-oxidation defects such as very long-chain fatty acid (VLCFA) accumulation. In these cell lines, we used RNA-sequencing and identified large-scale reprogramming for genes involved in lipid metabolism, immune response, cell signaling, lysosome and autophagy, as well as a DAM-like signature. We highlighted cholesterol accumulation in plasma membranes and observed autophagy patterns in the cell mutants. We confirmed the upregulation or downregulation at the protein level for a few selected genes that mostly corroborated our observations and clearly demonstrated increased expression and secretion of DAM proteins in the BV-2 mutant cells. In conclusion, the peroxisomal defects in microglial cells not only impact on VLCFA metabolism but also force microglial cells to adopt a pathological phenotype likely representing a key contributor to the pathogenesis of peroxisomal disorders.
Collapse
Affiliation(s)
- Quentin Raas
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| | - Ali Tawbeh
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| | - Mounia Tahri-Joutey
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences and Techniques, University Hassan I, Settat, Morocco
| | | | - Céline Keime
- Plateforme GenomEast, IGBMC, CNRS UMR 7104, Inserm U1258, University of Strasbourg, Illkirch, France
| | - Romain Kaiser
- Plateforme GenomEast, IGBMC, CNRS UMR 7104, Inserm U1258, University of Strasbourg, Illkirch, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| | - Boubker Nasser
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences and Techniques, University Hassan I, Settat, Morocco
| | - Valerio Leoni
- Laboratory of Clinical Biochemistry, Hospital of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Emma Bellanger
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Maud Boussand
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Yannick Hamon
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro Dijon, University of Bourgogne Franche-Comté, Dijon, France
| | - Francesca Di Cara
- Department of Microbiology and Immunology, IWK Health Centre, Dalhousie University, Halifax, NS, Canada
| | - Caroline Truntzer
- Platform of Transfer in Biological Oncology, Georges François Leclerc Cancer Center–Unicancer, Dijon, France
| | | | | | - Stéphane Savary
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
- *Correspondence: Stéphane Savary,
| |
Collapse
|
12
|
Ge MM, Chen N, Zhou YQ, Yang H, Tian YK, Ye DW. Galectin-3 in Microglia-Mediated Neuroinflammation: Implications for Central Nervous System Diseases. Curr Neuropharmacol 2022; 20:2066-2080. [PMID: 35105290 PMCID: PMC9886847 DOI: 10.2174/1570159x20666220201094547] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/27/2021] [Accepted: 01/29/2022] [Indexed: 11/22/2022] Open
Abstract
Microglial activation is one of the common hallmarks shared by various central nervous system (CNS) diseases. Based on surrounding circumstances, activated microglia play either detrimental or neuroprotective effects. Galectin-3 (Gal-3), a group of β-galactoside-binding proteins, has been cumulatively revealed to be a crucial biomarker for microglial activation after injuries or diseases. In consideration of the important role of Gal-3 in the regulation of microglial activation, it might be a potential target for the treatment of CNS diseases. Recently, Gal-3 expression has been extensively investigated in numerous pathological processes as a mediator of neuroinflammation, as well as in cell proliferation. However, the underlying mechanisms of Gal-3 involved in microgliamediated neuroinflammation in various CNS diseases remain to be further investigated. Moreover, several clinical studies support that the levels of Gal-3 are increased in the serum or cerebrospinal fluid of patients with CNS diseases. Thus, we summarized the roles and underlying mechanisms of Gal-3 in activated microglia, thus providing a better insight into its complexity expression pattern, and contrasting functions in CNS diseases.
Collapse
Affiliation(s)
- Meng-Meng Ge
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Nan Chen
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Hui Yang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Yu-Ke Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; ,Address correspondence to these authors at the Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. E-mail: ., Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. E-mail:
| | - Da-Wei Ye
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; ,Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China,Address correspondence to these authors at the Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. E-mail: ., Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. E-mail:
| |
Collapse
|
13
|
Galectins—Potential Therapeutic Targets for Neurodegenerative Disorders. Int J Mol Sci 2022; 23:ijms231911012. [PMID: 36232314 PMCID: PMC9569834 DOI: 10.3390/ijms231911012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Advancements in medicine have increased the longevity of humans, resulting in a higher incidence of chronic diseases. Due to the rise in the elderly population, age-dependent neurodegenerative disorders are becoming increasingly prevalent. The available treatment options only provide symptomatic relief and do not cure the underlying cause of the disease. Therefore, it has become imperative to discover new markers and therapies to modulate the course of disease progression and develop better treatment options for the affected individuals. Growing evidence indicates that neuroinflammation is a common factor and one of the main inducers of neuronal damage and degeneration. Galectins (Gals) are a class of β-galactoside-binding proteins (lectins) ubiquitously expressed in almost all vital organs. Gals modulate various cellular responses and regulate significant biological functions, including immune response, proliferation, differentiation, migration, and cell growth, through their interaction with glycoproteins and glycolipids. In recent years, extensive research has been conducted on the Gal superfamily, with Gal-1, Gal-3, and Gal-9 in prime focus. Their roles have been described in modulating neuroinflammation and neurodegenerative processes. In this review, we discuss the role of Gals in the causation and progression of neurodegenerative disorders. We describe the role of Gals in microglia and astrocyte modulation, along with their pro- and anti-inflammatory functions. In addition, we discuss the potential use of Gals as a novel therapeutic target for neuroinflammation and restoring tissue damage in neurodegenerative diseases.
Collapse
|
14
|
Galectin-3, a rising star in modulating microglia activation under conditions of neurodegeneration. Cell Death Dis 2022; 13:628. [PMID: 35859075 PMCID: PMC9300700 DOI: 10.1038/s41419-022-05058-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 01/21/2023]
Abstract
The advent of high-throughput single-cell transcriptomic analysis of microglia has revealed different phenotypes that are inherently associated with disease conditions. A common feature of some of these activated phenotypes is the upregulation of galectin-3. Representative examples of these phenotypes include disease-associated microglia (DAM) and white-associated microglia (WAM), whose role(s) in neuroprotection/neurotoxicity is a matter of high interest in the microglia community. In this review, we summarise the main findings that demonstrate the ability of galectin-3 to interact with key pattern recognition receptors, including, among others, TLR4 and TREM2 and the importance of galectin-3 in the regulation of microglia activation. Finally, we discuss increasing evidence supporting the involvement of this lectin in the main neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, traumatic brain injury, and stroke.
Collapse
|
15
|
Raffaele S, Fumagalli M. Dynamics of Microglia Activation in the Ischemic Brain: Implications for Myelin Repair and Functional Recovery. Front Cell Neurosci 2022; 16:950819. [PMID: 35899017 PMCID: PMC9309466 DOI: 10.3389/fncel.2022.950819] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is a neurological disorder representing a leading cause of death and permanent disability world-wide, for which effective regenerative treatments are missing. Oligodendrocyte degeneration and consequent myelin disruption are considered major contributing factors to stroke-associated neurological deficits. Therefore, fostering myelin reconstruction by oligodendrocyte precursor cells (OPCs) has emerged as a promising therapeutic approach to enhance functional recovery in stroke patients. A pivotal role in regulating remyelination is played by microglia, the resident immune cells of the brain. Early after stroke, microglial cells exert beneficial functions, promoting OPC recruitment toward the ischemic lesion and preserving myelin integrity. However, the protective features of microglia are lost during disease progression, contributing to remyelination failure. Unveiling the mechanisms driving the pro-remyelination properties of microglia may provide important opportunities for both reducing myelin damage and promoting its regeneration. Here, we summarize recent evidence describing microglia activation kinetics in experimental models of ischemic injury, focusing on the contribution of these innate immune cells to myelin damage and repair. Some molecular signals regulating the pro-regenerative functions of microglia after stroke have been highlighted to provide new possible therapeutic targets involved in the protective functions of these cells. Finally, we analyzed the impact of microglia-to-OPCs communication via extracellular vesicles on post-stroke remyelination and functional recovery. The results collected in this review underline the importance of supporting the pro-remyelination functions of microglial cells after stroke.
Collapse
|
16
|
Dupree JL, Paez PM, Tiwari-Woodruff SK, Denton TT, Hensley K, Angeliu CG, Boullerne AI, Kalinin S, Egge S, Cheli VT, Denaroso G, Atkinson KC, Feri M, Feinstein DL. Lanthionine Ketimine Ethyl Ester Accelerates Remyelination in a Mouse Model of Multiple Sclerosis. ASN Neuro 2022; 14:17590914221112352. [PMID: 35791633 PMCID: PMC9272172 DOI: 10.1177/17590914221112352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although over 20 disease modifying therapies are approved to treat Multiple Sclerosis (MS), these do not increase remyelination of demyelinated axons or mitigate axon damage. Previous studies showed that lanthionine ketenamine ethyl ester (LKE) reduces clinical signs in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS and increased maturation of oligodendrocyte (OL) progenitor cells (OPCs) in vitro. In the current study, we used the cuprizone (CPZ) demyelination model of MS to test if LKE could increase remyelination. The corpus callosum (CC) and somatosensory cortex was examined by immunohistochemistry (IHC), electron microscopy and for mRNA expression changes in mice provided 5 weeks of CPZ diet followed by 2 weeks of normal diet in the presence of LKE or vehicle. A significant increase in the number of myelinated axons, and increased myelin thickness was observed in the CC of LKE-treated groups compared to vehicle-treated groups. LKE also increased myelin basic protein and proteolipid protein expression in the CC and cortex, and increased the number of mature OLs in the cortex. In contrast, LKE did not increase the percentage of proliferating OPCs suggesting effects on OPC survival and differentiation but not proliferation. The effects of LKE on OL maturation and remyelination were supported by similar changes in their relative mRNA levels. Interestingly, LKE did not have significant effects on GFAP or Iba1 immunostaining or mRNA levels. These findings suggest that remyelinating actions of LKE can potentially be formulated to induce remyelination in neurological diseases associated with demyelination including MS.
Collapse
Affiliation(s)
- Jeffrey L. Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA,Research Service, HH McGuire VA Medical Center, Richmond, VA, USA
| | - Pablo M. Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | - Seema K. Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine at the University of California
Riverside, Riverside, CA, USA
| | - Travis T. Denton
- Department of Pharmaceutical Sciences, College of Pharmacy &
Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA,
USA,Department of Translational Medicine and Physiology, Elson S. Floyd College
of Medicine, Washington State University Health Sciences Spokane, Spokane, WA, USA,Steve Gleason Institute for Neuroscience, Washington State University Health Sciences
Spokane, Spokane, WA, USA
| | - Kenneth Hensley
- Arkansas College of Osteopathic
Medicine, Fort Smith, AR, USA
| | - Christina G. Angeliu
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | | | - Sergey Kalinin
- Department Anesthesiology, University of Illinois, Chicago, IL, USA
| | - Sophia Egge
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Veronica T. Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | - Giancarlo Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | - Kelley C. Atkinson
- Division of Biomedical Sciences, School of Medicine at the University of California
Riverside, Riverside, CA, USA
| | - Micah Feri
- Division of Biomedical Sciences, School of Medicine at the University of California
Riverside, Riverside, CA, USA
| | - Douglas L. Feinstein
- Department Anesthesiology, University of Illinois, Chicago, IL, USA,Jesse Brown VA Medical Center, Chicago, IL, USA,Douglas L. Feinstein, Department of Anesthesiology,
University of Illinois, 835 South Wolcott Avenue, MC 513, Chicago IL, 60612, USA.
| |
Collapse
|
17
|
Mijailović NR, Vesic K, Arsenijevic D, Milojević-Rakić M, Borovcanin MM. Galectin-3 Involvement in Cognitive Processes for New Therapeutic Considerations. Front Cell Neurosci 2022; 16:923811. [PMID: 35875353 PMCID: PMC9296991 DOI: 10.3389/fncel.2022.923811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Cognitive impairment may be a consequence of the normal aging process, but it may also be the hallmark of various neurodegenerative and psychiatric diseases. Early identification of individuals at particular risk for cognitive decline is critical, as it is imperative to maintain a cognitive reserve in these neuropsychiatric entities. In recent years, galectin-3 (Gal-3), a member of the galectin family, has received considerable attention with respect to aspects of neuroinflammation and neurodegeneration. The mechanisms behind the putative relationship between Gal-3 and cognitive impairment are not yet clear. Intrigued by this versatile molecule and its unique modular architecture, the latest data on this relationship are presented here. This mini-review summarizes recent findings on the mechanisms by which Gal-3 affects cognitive functioning in both animal and human models. Particular emphasis is placed on the role of Gal-3 in modulating the inflammatory response as a fine-tuner of microglia morphology and phenotype. A review of recent literature on the utility of Gal-3 as a biomarker is provided, and approaches to strategically exploit Gal-3 activities with therapeutic intentions in neuropsychiatric diseases are outlined.
Collapse
Affiliation(s)
- Nataša R. Mijailović
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- *Correspondence: Nataša R. Mijailović,
| | - Katarina Vesic
- Department of Neurology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Milica M. Borovcanin
- Department of Psychiatry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
18
|
Targeting microglia–oligodendrocyte crosstalk in neurodegenerative and psychiatric disorders. Drug Discov Today 2022; 27:2562-2573. [DOI: 10.1016/j.drudis.2022.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 06/09/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023]
|
19
|
The Inflammatory Response after Moderate Contusion Spinal Cord Injury: A Time Study. BIOLOGY 2022; 11:biology11060939. [PMID: 35741460 PMCID: PMC9220050 DOI: 10.3390/biology11060939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/30/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary The neuroinflammatory response is a rather complex event in spinal cord injury (SCI) and has the capacity to exacerbate cell damage but also to contribute to the repair of the injury. This complexity is thought to depend on a variety of inflammatory mediators, of which tumor necrosis factor (TNF) plays a key role. Evidence indicates that TNF can be both protective and detrimental in SCI. In the present study, we studied the temporal and cellular expression of TNF and its receptors after SCI in mice. We found TNF to be significantly increased in both the acute and the delayed phases after SCI, alongside a robust neuroinflammatory response. As we could verify some of our results in human postmortem tissue, our results imply that diminishing the detrimental immune signaling after SCI could also enhance recovery in humans. Abstract Spinal cord injury (SCI) initiates detrimental cellular and molecular events that lead to acute and delayed neuroinflammation. Understanding the role of the inflammatory response in SCI requires insight into the temporal and cellular synthesis of inflammatory mediators. We subjected C57BL/6J mice to SCI and investigated inflammatory reactions. We examined activation, recruitment, and polarization of microglia and infiltrating immune cells, focusing specifically on tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2. In the acute phase, TNF expression increased in glial cells and neuron-like cells, followed by infiltrating immune cells. TNFR1 and TNFR2 levels increased in the delayed phase and were found preferentially on neurons and glial cells, respectively. The acute phase was dominated by the infiltration of granulocytes and macrophages. Microglial/macrophage expression of Arg1 increased from 1–7 days after SCI, followed by an increase in Itgam, Cx3cr1, and P2ry12, which remained elevated throughout the study. By 21 and 28 days after SCI, the lesion core was populated by galectin-3+, CD68+, and CD11b+ microglia/macrophages, surrounded by a glial scar consisting of GFAP+ astrocytes. Findings were verified in postmortem tissue from individuals with SCI. Our findings support the consensus that future neuroprotective immunotherapies should aim to selectively neutralize detrimental immune signaling while sustaining pro-regenerative processes.
Collapse
|
20
|
Ramírez Hernández E, Alanis Olvera B, Carmona González D, Guerrero Marín O, Pantoja Mercado D, Valencia Gil L, Hernández-Zimbrón LF, Sánchez Salgado JL, Limón ID, Zenteno E. Neuroinflammation and galectins: a key relationship in neurodegenerative diseases. Glycoconj J 2022; 39:685-699. [PMID: 35653015 DOI: 10.1007/s10719-022-10064-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/16/2022]
Abstract
Neurodegeneration is a pathological condition that is associated with the loss of neuronal function and structure. In neurodegenerative diseases, mounting evidence indicates that neuroinflammation is a common factor that contributes to neuronal damage and neurodegeneration. Neuroinflammation is characterized by the activation of microglia, the neuroimmune cells of the central nervous system (CNS), which have been implicated as active contributors to neuronal damage. Glycan structure modification is defining the outcome of neuroinflammation and neuronal regeneration; moreover, the expression of galectins, a group of lectins that specifically recognize β-galactosides, has been proposed as a key factor in neuronal regeneration and modulation of the inflammatory response. Of the different galectins identified, galectin-1 stimulates the secretion of neurotrophic factors in astrocytes and promotes neuronal regeneration, whereas galectin-3 induces the proliferation of microglial cells and modulates cell apoptosis. Galectin-8 emerged as a neuroprotective factor, which, in addition to its immunosuppressive function, could generate a neuroprotective environment in the brain. This review describes the role of galectins in the activation and modulation of astrocytes and microglia and their anti- and proinflammatory functions within the context of neuroinflammation. Furthermore, it discusses the potential use of galectins as a therapeutic target for the inflammatory response and remodeling in damaged tissues in the central nervous system.
Collapse
Affiliation(s)
- Eleazar Ramírez Hernández
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | - Beatriz Alanis Olvera
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniela Carmona González
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Oscar Guerrero Marín
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Denisse Pantoja Mercado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lucero Valencia Gil
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis F Hernández-Zimbrón
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Luis Sánchez Salgado
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - I Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de México, Mexico City, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
21
|
Hohsfield LA, Tsourmas KI, Ghorbanian Y, Syage AR, Kim SJ, Cheng Y, Furman S, Inlay MA, Lane TE, Green KN. MAC2 is a long-lasting marker of peripheral cell infiltrates into the mouse CNS after bone marrow transplantation and coronavirus infection. Glia 2022; 70:875-891. [PMID: 35025109 PMCID: PMC8930563 DOI: 10.1002/glia.24144] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 01/09/2023]
Abstract
Microglia are the primary resident myeloid cells of the brain responsible for maintaining homeostasis and protecting the central nervous system (CNS) from damage and infection. Monocytes and monocyte-derived macrophages arising from the periphery have also been implicated in CNS pathologies, however, distinguishing between different myeloid cell populations in the CNS has been difficult. Here, we set out to develop a reliable histological marker that can assess distinct myeloid cell heterogeneity and functional contributions, particularly in the context of disease and/or neuroinflammation. scRNAseq from brains of mice infected with the neurotropic JHM strain of the mouse hepatitis virus (JHMV), a mouse coronavirus, revealed that Lgals3 is highly upregulated in monocyte and macrophage populations, but not in microglia. Subsequent immunostaining for galectin-3 (encoded by Lgals3), also referred to as MAC2, highlighted the high expression levels of MAC2 protein in infiltrating myeloid cells in JHMV-infected and bone marrow (BM) chimeric mice, in stark contrast to microglia, which expressed little to no staining in these models. Expression of MAC2 was found even 6-10 months following BM-derived cell infiltration into the CNS. We also demonstrate that MAC2 is not a specific label for plaque-associated microglia in the 5xFAD mouse model, but only appears in a distinct subset of these cells in the presence of JHMV infection or during aging. Our data suggest that MAC2 can serve as a reliable and long-lasting histological marker for monocyte/macrophages in the brain, identifying an accessible approach to distinguishing resident microglia from infiltrating cells in the CNS under certain conditions.
Collapse
Affiliation(s)
- Lindsay A. Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Kate Inman Tsourmas
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Yasamine Ghorbanian
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Amber R. Syage
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Sung Jin Kim
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Yuting Cheng
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Susana Furman
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Matthew A. Inlay
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Thomas E. Lane
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Kim N. Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| |
Collapse
|
22
|
Galectin-3: A Novel Marker for the Prediction of Stroke Incidence and Clinical Prognosis. Mediators Inflamm 2022; 2022:2924773. [PMID: 35281427 PMCID: PMC8904909 DOI: 10.1155/2022/2924773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 01/09/2022] [Accepted: 02/03/2022] [Indexed: 12/15/2022] Open
Abstract
Stroke, whether ischemic or haemorrhagic, is one of the main causes of mortality and disability all over the world, which entails huge burdens in both healthcare environments as well as social and economic aspects of life. Therefore, there is a continuous search for novel reliable biomarkers that can enhance the recognition of stroke events in a timely manner and predict the clinical outcomes following a stroke event. Galectins are a group of proteins expressed by many types of cells and tissues including vasculature, certain immune cells, fibroblasts, and gastrointestinal epithelial cells. These proteins vary in their structure and configuration according to their type and have a diversity of functions according to the type of tissue they are expressed in. Among these proteins, a few studies investigated mainly the roles played by galectin-1 (Gal-1) and galectin-3 (Gal-3) in the molecular mechanisms of atherosclerosis and in brain tissue remodeling after a stroke event. In this review, we present an updated overview of the current understanding of Gal-3's functions and implications in stroke occurrence and the response of the brain tissue to stroke events, which may be a key to its utility as a predictor of stroke incidence and clinical prognosis in the future.
Collapse
|
23
|
Salazar IL, Lourenço AST, Manadas B, Baldeiras I, Ferreira C, Teixeira AC, Mendes VM, Novo AM, Machado R, Batista S, Macário MDC, Grãos M, Sousa L, Saraiva MJ, Pais AACC, Duarte CB. Posttranslational modifications of proteins are key features in the identification of CSF biomarkers of multiple sclerosis. J Neuroinflammation 2022; 19:44. [PMID: 35135578 PMCID: PMC8822857 DOI: 10.1186/s12974-022-02404-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/26/2022] [Indexed: 12/27/2022] Open
Abstract
Background Multiple sclerosis is an inflammatory and degenerative disease of the central nervous system (CNS) characterized by demyelination and concomitant axonal loss. The lack of a single specific test, and the similarity to other inflammatory diseases of the central nervous system, makes it difficult to have a clear diagnosis of multiple sclerosis. Therefore, laboratory tests that allows a clear and definite diagnosis, as well as to predict the different clinical courses of the disease are of utmost importance. Herein, we compared the cerebrospinal fluid (CSF) proteome of patients with multiple sclerosis (in the relapse–remitting phase of the disease) and other diseases of the CNS (inflammatory and non-inflammatory) aiming at identifying reliable biomarkers of multiple sclerosis. Methods CSF samples from the discovery group were resolved by 2D-gel electrophoresis followed by identification of the protein spots by mass spectrometry. The results were analyzed using univariate (Student’s t test) and multivariate (Hierarchical Cluster Analysis, Principal Component Analysis, Linear Discriminant Analysis) statistical and numerical techniques, to identify a set of protein spots that were differentially expressed in CSF samples from patients with multiple sclerosis when compared with other two groups. Validation of the results was performed in samples from a different set of patients using quantitative (e.g., ELISA) and semi-quantitative (e.g., Western Blot) experimental approaches. Results Analysis of the 2D-gels showed 13 protein spots that were differentially expressed in the three groups of patients: Alpha-1-antichymotrypsin, Prostaglandin-H2-isomerase, Retinol binding protein 4, Transthyretin (TTR), Apolipoprotein E, Gelsolin, Angiotensinogen, Agrin, Serum albumin, Myosin-15, Apolipoprotein B-100 and EF-hand calcium-binding domain—containing protein. ELISA experiments allowed validating part of the results obtained in the proteomics analysis and showed that some of the alterations in the CSF proteome are also mirrored in serum samples from multiple sclerosis patients. CSF of multiple sclerosis patients was characterized by TTR oligomerization, thus highlighting the importance of analyzing posttranslational modifications of the proteome in the identification of novel biomarkers of the disease. Conclusions The model built based on the results obtained upon analysis of the 2D-gels and in the validation phase attained an accuracy of about 80% in distinguishing multiple sclerosis patients and the other two groups. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02404-2.
Collapse
Affiliation(s)
- Ivan L Salazar
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Ana S T Lourenço
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cláudia Ferreira
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Anabela Claro Teixeira
- Molecular Neurobiology Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Vera M Mendes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana Margarida Novo
- Neurology Department, CHUC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Rita Machado
- Neurology Department, CHUC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sónia Batista
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Neurology Department, CHUC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Maria do Carmo Macário
- Neurology Department, CHUC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Mário Grãos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.,Biocant-Associação de Transferência de Tecnologia, Cantanhede, Portugal
| | - Lívia Sousa
- Neurology Department, CHUC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Maria João Saraiva
- Molecular Neurobiology Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Alberto A C C Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | - Carlos B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
24
|
Alterations of Plasma Galectin-3 and C3 Levels in Patients with Parkinson's Disease. Brain Sci 2021; 11:brainsci11111515. [PMID: 34827514 PMCID: PMC8615685 DOI: 10.3390/brainsci11111515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 12/03/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by progressive neurodegeneration of dopaminergic neurons in the ventral midbrain. The complement-phagosome pathway is involved in the pathogenesis of PD. Here we measured levels of complement-phagocytosis molecules, including galectin-3, C3, C4, and cathepsin D, in the plasma of 56 patients with PD, and 46 normal controls (NCs). Plasma levels of galectin-3 (9.93 ± 3.94 ng/mL) were significantly higher in PD patients compared with NCs (8.39 ± 1.95 ng/mL, p = 0.012), and demonstrated a positive correlation with Hoehn and Yahr stages in PD patients (R2 = 0.218, p < 0.001). On the other hand, plasma C3 levels were significantly lower in PD patients (305.27 ± 205.16 μg/mL) compared with NCs (444.34 ± 245.54 μg/mL, p = 0.002). However, the levels did not correlate with Hoehn and Yahr stages (R2 = 0.010, p = 0.469). Plasma levels of C4 and cathepsin D in PD patients were similar to those in NCs. Our results show possible altered complement-phagocytosis signals in the peripheral blood of PD patients, highlighting the potential of galectin-3 as a biomarker of PD.
Collapse
|
25
|
Nio-Kobayashi J, Itabashi T. Galectins and Their Ligand Glycoconjugates in the Central Nervous System Under Physiological and Pathological Conditions. Front Neuroanat 2021; 15:767330. [PMID: 34720894 PMCID: PMC8554236 DOI: 10.3389/fnana.2021.767330] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Galectins are β-galactoside-binding lectins consisting of 15 members in mammals. Galectin-1,-3,-4,-8, and -9 are predominantly expressed in the central nervous system (CNS) and regulate various physiological and pathological events. This review summarizes the current knowledge of the cellular expression and role of galectins in the CNS, and discusses their functions in neurite outgrowth, myelination, and neural stem/progenitor cell niches, as well as in ischemic/hypoxic/traumatic injuries and neurodegenerative diseases such as multiple sclerosis. Galectins are expressed in both neurons and glial cells. Galectin-1 is mainly expressed in motoneurons, whereas galectin-3-positive neurons are broadly distributed throughout the brain, especially in the hypothalamus, indicating its function in the regulation of homeostasis, stress response, and the endocrine/autonomic system. Astrocytes predominantly contain galectin-1, and galectin-3 and−9 are upregulated along with its activation. Activated, but not resting, microglia contain galectin-3, supporting its phagocytic activity. Galectin-1,−3, and -4 are characteristically expressed during oligodendrocyte differentiation. Galectin-3 from microglia promotes oligodendrocyte differentiation and myelination, while galectin-1 and axonal galectin-4 suppress its differentiation and myelination. Galectin-1- and- 3-positive cells are involved in neural stem cell niche formation in the subventricular zone and hippocampal dentate gyrus, and the migration of newly generated neurons and glial cells to the olfactory bulb or damaged lesions. In neurodegenerative diseases, galectin-1,-8, and -9 have neuroprotective and anti-inflammatory activities. Galectin-3 facilitates pro-inflammatory action; however, it also plays an important role during the recovery period. Several ligand glycoconjugates have been identified so far such as laminin, integrins, neural cell adhesion molecule L1, sulfatide, neuropilin-1/plexinA4 receptor complex, triggering receptor on myeloid cells 2, and T cell immunoglobulin and mucin domain. N-glycan branching on lymphocytes and oligodendroglial progenitors mediated by β1,6-N-acetylglucosaminyltransferase V (Mgat5/GnTV) influences galectin-binding, modulating inflammatory responses and remyelination in neurodegenerative diseases. De-sulfated galactosaminoglycans such as keratan sulfate are potential ligands for galectins, especially galectin-3, regulating neural regeneration. Galectins have multitudinous functions depending on cell type and context as well as post-translational modifications, including oxidization, phosphorylation, S-nitrosylation, and cleavage, but there should be certain rules in the expression patterns of galectins and their ligand glycoconjugates, possibly related to glucose metabolism in cells.
Collapse
Affiliation(s)
- Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tetsuya Itabashi
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
26
|
Sørensen MD, Kristensen BW. TUMOUR-ASSOCIATED CD204+ MICROGLIA/MACROPHAGES ACCUMULATE IN PERIVASCULAR AND PERINECROTIC NICHES AND CORRELATE WITH AN INTERLEUKIN-6 ENRICHED INFLAMMATORY PROFILE IN GLIOBLASTOMA. Neuropathol Appl Neurobiol 2021; 48:e12772. [PMID: 34713474 PMCID: PMC9306597 DOI: 10.1111/nan.12772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Mia Dahl Sørensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Lee MJ, Wang C, Carroll MJ, Brubaker DK, Hyman BT, Lauffenburger DA. Computational Interspecies Translation Between Alzheimer's Disease Mouse Models and Human Subjects Identifies Innate Immune Complement, TYROBP, and TAM Receptor Agonist Signatures, Distinct From Influences of Aging. Front Neurosci 2021; 15:727784. [PMID: 34658769 PMCID: PMC8515135 DOI: 10.3389/fnins.2021.727784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 11/30/2022] Open
Abstract
Mouse models are vital for preclinical research on Alzheimer’s disease (AD) pathobiology. Many traditional models are driven by autosomal dominant mutations identified from early onset AD genetics whereas late onset and sporadic forms of the disease are predominant among human patients. Alongside ongoing experimental efforts to improve fidelity of mouse model representation of late onset AD, a computational framework termed Translatable Components Regression (TransComp-R) offers a complementary approach to leverage human and mouse datasets concurrently to enhance translation capabilities. We employ TransComp-R to integratively analyze transcriptomic data from human postmortem and traditional amyloid mouse model hippocampi to identify pathway-level signatures present in human patient samples yet predictive of mouse model disease status. This method allows concomitant evaluation of datasets across different species beyond observational seeking of direct commonalities between the species. Additional linear modeling focuses on decoupling disease signatures from effects of aging. Our results elucidated mouse-to-human translatable signatures associated with disease: excitatory synapses, inflammatory cytokine signaling, and complement cascade- and TYROBP-based innate immune activity; these signatures all find validation in previous literature. Additionally, we identified agonists of the Tyro3 / Axl / MerTK (TAM) receptor family as significant contributors to the cross-species innate immune signature; the mechanistic roles of the TAM receptor family in AD merit further dedicated study. We have demonstrated that TransComp-R can enhance translational understanding of relationships between AD mouse model data and human data, thus aiding generation of biological hypotheses concerning AD progression and holding promise for improved preclinical evaluation of therapies.
Collapse
Affiliation(s)
- Meelim J Lee
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Molly J Carroll
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Douglas K Brubaker
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN, United States
| | - Bradley T Hyman
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, United States
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
28
|
Borovcanin MM, Vesic K, Jovanovic M, Mijailovic NR. Galectin-3 possible involvement in antipsychotic-induced metabolic changes of schizophrenia: A minireview. World J Diabetes 2021; 12:1731-1739. [PMID: 34754374 PMCID: PMC8554363 DOI: 10.4239/wjd.v12.i10.1731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/24/2021] [Accepted: 08/06/2021] [Indexed: 02/06/2023] Open
Abstract
Recently, specific immunometabolic profiles have been postulated in patients with schizophrenia, even before full-blown disease and independent of antipsychotic treatment. Proteomic profiling studies offer a promising potential for elucidating the cellular and molecular pathways that may be involved in the onset and progression of schizophrenia symptoms, and co-occurrent metabolic changes. In view of all this, we were intrigued to explore galectin-3 (Gal-3) as a glycan, and in our previous study, we measured its elevated levels in remission of schizophrenia. The finding may be a consequence of antipsychotic treatment and may have an impact on the onset of inflammation, the development of obesity, and the presumed cognitive changes in schizophrenia. In the animal study, it was shown that downregulation of Gal-3 was beneficial in insulin regulation of obesity and cognitive preservation. Strategies involving plasma exchange are discussed in this review, particularly in the context of Gal-3 elimination.
Collapse
Affiliation(s)
- Milica M Borovcanin
- Department of Psychiatry, University of Kragujevac, Faculty of Medical Sciences, Kragujevac 34000, Sumadija, Serbia
| | - Katarina Vesic
- Department of Neurology, University of Kragujevac, Faculty of Medical Sciences, Kragujevac 34000, Sumadija, Serbia
| | - Milena Jovanovic
- PhD Studies, University of Kragujevac, Faculty of Medical Sciences, Kragujevac 34000, Sumadija, Serbia
- Clinic for Nephrology and Dialysis, University Clinical Center Kragujevac, Kragujevac 34000, Sumadija, Serbia
| | - Natasa R Mijailovic
- Department of Pharmacy, University of Kragujevac, Faculty of Medical Sciences, Kragujevac 34000, Sumadija, Serbia
| |
Collapse
|
29
|
Rahimian R, Wakid M, O'Leary LA, Mechawar N. The emerging tale of microglia in psychiatric disorders. Neurosci Biobehav Rev 2021; 131:1-29. [PMID: 34536460 DOI: 10.1016/j.neubiorev.2021.09.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
As the professional phagocytes of the brain, microglia orchestrate the immunological response and play an increasingly important role in maintaining homeostatic brain functions. Microglia are activated by pathological events or slight alterations in brain homeostasis. This activation is dependent on the context and type of stressor or pathology. Through secretion of cytokines, chemokines and growth factors, microglia can strongly influence the response to a stressor and can, therefore, determine the pathological outcome. Psychopathologies have repeatedly been associated with long-lasting priming and sensitization of cerebral microglia. This review focuses on the diversity of microglial phenotype and function in health and psychiatric disease. We first discuss the diverse homeostatic functions performed by microglia and then elaborate on context-specific spatial and temporal microglial heterogeneity. Subsequently, we summarize microglia involvement in psychopathologies, namely major depressive disorder, schizophrenia and bipolar disorder, with a particular focus on post-mortem studies. Finally, we postulate microglia as a promising novel therapeutic target in psychiatry through antidepressant and antipsychotic treatment.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada
| | - Marina Wakid
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Liam Anuj O'Leary
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
30
|
Psenicka MW, Smith BC, Tinkey RA, Williams JL. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? Front Cell Neurosci 2021; 15:654284. [PMID: 34234647 PMCID: PMC8255483 DOI: 10.3389/fncel.2021.654284] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The pathology in neurodegenerative diseases is often accompanied by inflammation. It is well-known that many cells within the central nervous system (CNS) also contribute to ongoing neuroinflammation, which can promote neurodegeneration. Multiple sclerosis (MS) is both an inflammatory and neurodegenerative disease in which there is a complex interplay between resident CNS cells to mediate myelin and axonal damage, and this communication network can vary depending on the subtype and chronicity of disease. Oligodendrocytes, the myelinating cell of the CNS, and their precursors, oligodendrocyte precursor cells (OPCs), are often thought of as the targets of autoimmune pathology during MS and in several animal models of MS; however, there is emerging evidence that OPCs actively contribute to inflammation that directly and indirectly contributes to neurodegeneration. Here we discuss several contributors to MS disease progression starting with lesion pathology and murine models amenable to studying particular aspects of disease. We then review how OPCs themselves can play an active role in promoting neuroinflammation and neurodegeneration, and how other resident CNS cells including microglia, astrocytes, and neurons can impact OPC function. Further, we outline the very complex and pleiotropic role(s) of several inflammatory cytokines and other secreted factors classically described as solely deleterious during MS and its animal models, but in fact, have many neuroprotective functions and promote a return to homeostasis, in part via modulation of OPC function. Finally, since MS affects patients from the onset of disease throughout their lifespan, we discuss the impact of aging on OPC function and CNS recovery. It is becoming clear that OPCs are not simply a bystander during MS progression and uncovering the active roles they play during different stages of disease will help uncover potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Morgan W. Psenicka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| |
Collapse
|
31
|
Caridi B, Doncheva D, Sivaprasad S, Turowski P. Galectins in the Pathogenesis of Common Retinal Disease. Front Pharmacol 2021; 12:687495. [PMID: 34079467 PMCID: PMC8165321 DOI: 10.3389/fphar.2021.687495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Diseases of the retina are major causes of visual impairment and blindness in developed countries and, due to an ageing population, their prevalence is continually rising. The lack of effective therapies and the limitations of those currently in use highlight the importance of continued research into the pathogenesis of these diseases. Vascular endothelial growth factor (VEGF) plays a major role in driving vascular dysfunction in retinal disease and has therefore become a key therapeutic target. Recent evidence also points to a potentially similarly important role of galectins, a family of β-galactoside-binding proteins. Indeed, they have been implicated in regulating fundamental processes, including vascular hyperpermeability, angiogenesis, neuroinflammation, and oxidative stress, all of which also play a prominent role in retinopathies. Here, we review direct evidence for pathological roles of galectins in retinal disease. In addition, we extrapolate potential roles of galectins in the retina from evidence in cancer, immune and neuro-biology. We conclude that there is value in increasing understanding of galectin function in retinal biology, in particular in the context of the retinal vasculature and microglia. With greater insight, recent clinical developments of galectin-targeting drugs could potentially also be of benefit to the clinical management of many blinding diseases.
Collapse
Affiliation(s)
- Bruna Caridi
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Dilyana Doncheva
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Sobha Sivaprasad
- UCL Institute of Ophthalmology, University College London, London, United Kingdom.,NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
32
|
Zhou L, Matsushima GK. Tyro3, Axl, Mertk receptor-mediated efferocytosis and immune regulation in the tumor environment. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 361:165-210. [PMID: 34074493 DOI: 10.1016/bs.ircmb.2021.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Three structurally related tyrosine receptor cell surface kinases, Tyro3, Axl, and Mertk (TAM) have been recognized to modulate immune function, tissue homeostasis, cardiovasculature, and cancer. The TAM receptor family appears to operate in adult mammals across multiple cell types, suggesting both widespread and specific regulation of cell functions and immune niches. TAM family members regulate tissue homeostasis by monitoring the presence of phosphatidylserine expressed on stressed or apoptotic cells. The detection of phosphatidylserine on apoptotic cells requires intermediary molecules that opsonize the dying cells and tether them to TAM receptors on phagocytes. This complex promotes the engulfment of apoptotic cells, also known as efferocytosis, that leads to the resolution of inflammation and tissue healing. The immune mechanisms dictating these processes appear to fall upon specific family members or may involve a complex of different receptors acting cooperatively to resolve and repair damaged tissues. Here, we focus on the role of TAM receptors in triggering efferocytosis and its consequences in the regulation of immune responses in the context of inflammation and cancer.
Collapse
Affiliation(s)
- Liwen Zhou
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC, United States
| | - Glenn K Matsushima
- UNC Neuroscience Center, University of North Carolina-CH, Chapel Hill, NC, United States; UNC Department of Microbiology & Immunology, University of North Carolina-CH, Chapel Hill, NC, United States; UNC Integrative Program for Biological & Genome Sciences, University of North Carolina-CH, Chapel Hill, NC, United States.
| |
Collapse
|
33
|
Srejovic IM, Lukic ML. Galectin-3 in T cell-mediated immunopathology and autoimmunity. Immunol Lett 2021; 233:57-67. [PMID: 33753135 DOI: 10.1016/j.imlet.2021.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/17/2021] [Indexed: 01/05/2023]
Abstract
Galectin-3 (Gal-3) is the only member of galectin family able to form pentamers and heterodimers with chemokines. Its presence in various cells and tissues suggests variety of regulatory functions in physiological conditions, but increasing body of evidence indicates involvement of Gal-3 in pathological cascades of many diseases. Gal-3 exerts different, sometimes opposite, effects in various disorders or in different phases of the same disease. These differences in action of Gal-3 are related to the localization of Gal-3 in the cell, types of receptors through which it acts, or the types of cells that secrete it. As a regulator of immune response and T-cell activity, Gal-3 appears to have important role in development of autoimmunity mediated by T cells. Absence of Gal-3 in C57Bl6 mice favors Th2 mediated inflammatory myocarditis but attenuate fibrosis. Recent data also indicate Gal-3 involvement in development atherosclerosis. In pathogenesis of diabetes type 1 and autoimmune components of diabetes type 2 Gal-3 may have detrimental or protective role depending on its intracellular or extracellular localization. Gal-3 mediates autoimmune hepatic damage through activation of T-cells or natural killer T cells. Gal-3 is an important mediator in neurodevelopment, neuropathology and behavior due to its expression both in neurons and glial cells. All together, assessing the role of Gal-3 in immunopathology and autoimmunity it could be concluded that it is an important participant in pathogenesis, as well as promising monitoring marker and therapeutic target.
Collapse
Affiliation(s)
- Ivan M Srejovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000, Kragujevac, Serbia.
| | - Miodrag L Lukic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000, Kragujevac, Serbia; University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, Svetozara Markovica 69, 34000, Kragujevac, Serbia.
| |
Collapse
|
34
|
Rahimian R, Béland LC, Sato S, Kriz J. Microglia-derived galectin-3 in neuroinflammation; a bittersweet ligand? Med Res Rev 2021; 41:2582-2589. [PMID: 33733487 DOI: 10.1002/med.21784] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/24/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022]
Abstract
Galectins are soluble β-galactoside-binding proteins found in all multicellular organisms. Galectins may act as danger-associated molecular patterns in innate immunity and/or as pattern-recognition receptors that bind to pathogen-associated molecular patterns. Among different galectin family members, galectin-3 has been the focus of studies in neurodegenerative diseases in recent years. This lectin modulates brain innate immune responses, microglia activation patterns in physiological and pathophysiological settings in a context-dependent manner. Galectin-3 is considered as a pivotal tuner of macrophage and microglial activity. Indeed galectin-3 acts as a double edged sword in neuroinflammatory context and this multimodal lectin has diverse roles in physiological and pathophysiological conditions. Better understanding of galectin-3 physiology (its extracellular and intracellular actions) and structure (its C terminus vs. N terminus) is instrumental to design molecules that selectively modulate galectin-3 function toward neuroprotective phenotypes. Several experimental studies using different approaches and methods have demonstrated both protective and deleterious effects of galectin-3 in neuroinflammatory diseases. According to the crucial role of galectin-3 in modulation of innate immune response in brain, it is an attractive target in drug discovery of neurodegenerative diseases. The current insight attempts to provide an updated and balanced discussion on the role of galectin-3 as a complex endogenous immune modulator. This helps to have a better insight into the development of galectin-3 modulators with translational value in different neurological disorders including stroke and neurodegenerative diseases, such as Alzheimer's disease, Huntington's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, McGill University, Montreal, Quebec, Canada
| | | | - Sachiko Sato
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases, CHU de Quebec Research Centre, Faculty of Medicine, Laval University, Quebec, Quebec, Canada
| | - Jasna Kriz
- CERVO Brain Research Centre and Department of Psychiatry and Neuroscience, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
35
|
Loss of NPC1 enhances phagocytic uptake and impairs lipid trafficking in microglia. Nat Commun 2021; 12:1158. [PMID: 33627648 PMCID: PMC7904859 DOI: 10.1038/s41467-021-21428-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Niemann-Pick type C disease is a rare neurodegenerative disorder mainly caused by mutations in NPC1, resulting in abnormal late endosomal/lysosomal lipid storage. Although microgliosis is a prominent pathological feature, direct consequences of NPC1 loss on microglial function remain not fully characterized. We discovered pathological proteomic signatures and phenotypes in NPC1-deficient murine models and demonstrate a cell autonomous function of NPC1 in microglia. Loss of NPC1 triggers enhanced phagocytic uptake and impaired myelin turnover in microglia that precede neuronal death. Npc1−/− microglia feature a striking accumulation of multivesicular bodies and impaired trafficking of lipids to lysosomes while lysosomal degradation function remains preserved. Molecular and functional defects were also detected in blood-derived macrophages of NPC patients that provide a potential tool for monitoring disease. Our study underscores an essential cell autonomous role for NPC1 in immune cells and implies microglial therapeutic potential. Niemann-Pick type C disease is a rare childhood neurodegenerative disorder predominantly caused by mutations in NPC1, resulting in abnormal late endosomal and lysosomal defects. Here the authors show that NPC1 disruption largely impairs microglial function.
Collapse
|
36
|
Pasquini LA. Novel Galectin-3 interactions involved in oligodendroglial differentiation make inroads into therapeutic strategies for demyelinating diseases. Neural Regen Res 2021; 16:289-290. [PMID: 32859779 PMCID: PMC7896232 DOI: 10.4103/1673-5374.290887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Laura Andrea Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Biological Chemistry and Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
37
|
Ramakrishnan P. Could Galectin-3 be a key player in the etiology of neuromyelitis optica spectrum disorder? Med Hypotheses 2020; 146:110450. [PMID: 33309338 DOI: 10.1016/j.mehy.2020.110450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022]
Abstract
Neuromyelitis Optica Spectrum Disorder (NMOSD) is a chronic, inflammatory, demyelinating disorder of the central nervous system (CNS) characterized primarily by transverse myelitis (TM) and optic neuritis (ON). Serum antibodies of the IgG class to the water channel protein aquaporin-4 (AQP4) are associated with NMOSD in most cases. These antibodies are thought to cause functional abnormality or changed expressional pattern of AQP4 channel proteins in the CNS lesions. Activation of microglia is one of the chief antibody-mediated effects in NMOSD and it has opposing detrimental and protective effects in NMOSD. On the one hand, it promotes neuroinflammation, demyelination and BBB breakdown. On the other, it aids in remyelination. What controls the switch between these effects is unknown. Recently, Galectin- 3, a lectin, has been identified as a key player in several neurodegenerative diseases. In transient focal brain ischemia, alzheimer's disease (AD), huntington disease (HD), and experimental autoimmune encephalitis (EAE), Galectin-3 promotes microglia-mediated inflammation. Conversely, in amyotrophic lateral sclerosis (ALS), Galectin-3 reduces inflammation. It also suppresses Th17 cytokines, which play a crucial role in NMOSD pathogenesis. Being devoid of a leader signal, Gal-3 localizes in different cellular compartments and is subject to various post-translational modifications. These reasons explain why Galectin-3 expression has opposing effects under different physiological conditions. Microglia-mediated inflammation in NMOSD has not been extensively studied. The factors that regulate microglia-mediated inflammation in NMOSD are unknown. Here, I hypothesize that Galectin-3 might be an etiological factor in NMOSD that regulates microglia-mediated inflammation. Analysing the role of Gal-3 in NMOSD could help in the development of novel therapies to treat NMOSD.
Collapse
|
38
|
Mangale V, Syage AR, Ekiz HA, Skinner DD, Cheng Y, Stone CL, Brown RM, O'Connell RM, Green KN, Lane TE. Microglia influence host defense, disease, and repair following murine coronavirus infection of the central nervous system. Glia 2020; 68:2345-2360. [PMID: 32449994 PMCID: PMC7280614 DOI: 10.1002/glia.23844] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022]
Abstract
The present study examines functional contributions of microglia in host defense, demyelination, and remyelination following infection of susceptible mice with a neurotropic coronavirus. Treatment with PLX5622, an inhibitor of colony stimulating factor 1 receptor (CSF1R) that efficiently depletes microglia, prior to infection of the central nervous system (CNS) with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in increased mortality compared with control mice that correlated with impaired control of viral replication. Single cell RNA sequencing (scRNASeq) of CD45+ cells isolated from the CNS revealed that PLX5622 treatment resulted in muted CD4+ T cell activation profile that was associated with decreased expression of transcripts encoding MHC class II and CD86 in macrophages but not dendritic cells. Evaluation of spinal cord demyelination revealed a marked increase in white matter damage in PLX5622-treated mice that corresponded with elevated expression of transcripts encoding disease-associated proteins Osteopontin (Spp1), Apolipoprotein E (Apoe), and Triggering receptor expressed on myeloid cells 2 (Trem2) that were enriched within macrophages. In addition, PLX5622 treatment dampened expression of Cystatin F (Cst7), Insulin growth factor 1 (Igf1), and lipoprotein lipase (Lpl) within macrophage populations which have been implicated in promoting repair of damaged nerve tissue and this was associated with impaired remyelination. Collectively, these findings argue that microglia tailor the CNS microenvironment to enhance control of coronavirus replication as well as dampen the severity of demyelination and influence repair.
Collapse
Affiliation(s)
- Vrushali Mangale
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Amber R. Syage
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - H. Atakan Ekiz
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Dominic D. Skinner
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Yuting Cheng
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Colleen L. Stone
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - R. Marshall Brown
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Ryan M. O'Connell
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Kim N. Green
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
39
|
Huang M, Modeste E, Dammer E, Merino P, Taylor G, Duong DM, Deng Q, Holler CJ, Gearing M, Dickson D, Seyfried NT, Kukar T. Network analysis of the progranulin-deficient mouse brain proteome reveals pathogenic mechanisms shared in human frontotemporal dementia caused by GRN mutations. Acta Neuropathol Commun 2020; 8:163. [PMID: 33028409 PMCID: PMC7541308 DOI: 10.1186/s40478-020-01037-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 02/08/2023] Open
Abstract
Heterozygous, loss-of-function mutations in the granulin gene (GRN) encoding progranulin (PGRN) are a common cause of frontotemporal dementia (FTD). Homozygous GRN mutations cause neuronal ceroid lipofuscinosis-11 (CLN11), a lysosome storage disease. PGRN is a secreted glycoprotein that can be proteolytically cleaved into seven bioactive 6 kDa granulins. However, it is unclear how deficiency of PGRN and granulins causes neurodegeneration. To gain insight into the mechanisms of FTD pathogenesis, we utilized Tandem Mass Tag isobaric labeling mass spectrometry to perform an unbiased quantitative proteomic analysis of whole-brain tissue from wild type (Grn+/+) and Grn knockout (Grn-/-) mice at 3- and 19-months of age. At 3-months lysosomal proteins (i.e. Gns, Scarb2, Hexb) are selectively increased indicating lysosomal dysfunction is an early consequence of PGRN deficiency. Additionally, proteins involved in lipid metabolism (Acly, Apoc3, Asah1, Gpld1, Ppt1, and Naaa) are decreased; suggesting lysosomal degradation of lipids may be impaired in the Grn-/- brain. Systems biology using weighted correlation network analysis (WGCNA) of the Grn-/- brain proteome identified 26 modules of highly co-expressed proteins. Three modules strongly correlated to Grn deficiency and were enriched with lysosomal proteins (Gpnmb, CtsD, CtsZ, and Tpp1) and inflammatory proteins (Lgals3, GFAP, CD44, S100a, and C1qa). We find that lysosomal dysregulation is exacerbated with age in the Grn-/- mouse brain leading to neuroinflammation, synaptic loss, and decreased markers of oligodendrocytes, myelin, and neurons. In particular, GPNMB and LGALS3 (galectin-3) were upregulated by microglia and elevated in FTD-GRN brain samples, indicating common pathogenic pathways are dysregulated in human FTD cases and Grn-/- mice. GPNMB levels were significantly increased in the cerebrospinal fluid of FTD-GRN patients, but not in MAPT or C9orf72 carriers, suggesting GPNMB could be a biomarker specific to FTD-GRN to monitor disease onset, progression, and drug response. Our findings support the idea that insufficiency of PGRN and granulins in humans causes neurodegeneration through lysosomal dysfunction, defects in autophagy, and neuroinflammation, which could be targeted to develop effective therapies.
Collapse
|
40
|
Sariol A, Mackin S, Allred MG, Ma C, Zhou Y, Zhang Q, Zou X, Abrahante JE, Meyerholz DK, Perlman S. Microglia depletion exacerbates demyelination and impairs remyelination in a neurotropic coronavirus infection. Proc Natl Acad Sci U S A 2020; 117:24464-24474. [PMID: 32929007 PMCID: PMC7533697 DOI: 10.1073/pnas.2007814117] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microglia are considered both pathogenic and protective during recovery from demyelination, but their precise role remains ill defined. Here, using an inhibitor of colony stimulating factor 1 receptor (CSF1R), PLX5622, and mice infected with a neurotropic coronavirus (mouse hepatitis virus [MHV], strain JHMV), we show that depletion of microglia during the time of JHMV clearance resulted in impaired myelin repair and prolonged clinical disease without affecting the kinetics of virus clearance. Microglia were required only during the early stages of remyelination. Notably, large deposits of extracellular vesiculated myelin and cellular debris were detected in the spinal cords of PLX5622-treated and not control mice, which correlated with decreased numbers of oligodendrocytes in demyelinating lesions in drug-treated mice. Furthermore, gene expression analyses demonstrated differential expression of genes involved in myelin debris clearance, lipid and cholesterol recycling, and promotion of oligodendrocyte function. The results also demonstrate that microglial functions affected by depletion could not be compensated by infiltrating macrophages. Together, these results demonstrate that microglia play key roles in debris clearance and in the initiation of remyelination following infection with a neurotropic coronavirus but are not necessary during later stages of remyelination.
Collapse
Affiliation(s)
- Alan Sariol
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Samantha Mackin
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| | - Merri-Grace Allred
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Chen Ma
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Yu Zhou
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Qinran Zhang
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Juan E Abrahante
- University of Minnesota Informatics Institute (UMII), Minneapolis, MN 55455
| | | | - Stanley Perlman
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
41
|
Pascua-Maestro R, Corraliza-Gomez M, Fadrique-Rojo C, Ledesma MD, Schuchman EH, Sanchez D, Ganfornina MD. Apolipoprotein D-mediated preservation of lysosomal function promotes cell survival and delays motor impairment in Niemann-Pick type A disease. Neurobiol Dis 2020; 144:105046. [PMID: 32798728 DOI: 10.1016/j.nbd.2020.105046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 12/31/2022] Open
Abstract
Lysosomal Storage Diseases (LSD) are genetic diseases causing systemic and nervous system dysfunction. The glia-derived lipid binding protein Apolipoprotein D (ApoD) is required for lysosomal functional integrity in glial and neuronal cells, ensuring cell survival upon oxidative stress or injury. Here we test whether ApoD counteracts the pathogenic consequences of a LSD, Niemann Pick-type-A disease (NPA), where mutations in the acid sphingomyelinase gene result in sphingomyelin accumulation, lysosomal permeabilization and early-onset neurodegeneration. We performed a multivariable analysis of behavioral, cellular and molecular outputs in 12 and 24 week-old male and female NPA model mice, combined with ApoD loss-of-function mutation. Lack of ApoD in NPA mice accelerates cerebellar-dependent motor deficits, enhancing loss of Purkinje neurons. We studied ApoD expression in brain sections from a NPA patient and age-matched control, and the functional consequences of ApoD supplementation in primary human fibroblasts from two independent NPA patients and two control subjects. Cell viability, lipid peroxidation, and lysosomal functional integrity (pH, Cathepsin B activity, Galectin-3 exclusion) were examined. ApoD is endogenously overexpressed in NPA patients and NPA mouse brains and targeted to lysosomes of NPA patient cells, including Purkinje neurons and cultured fibroblasts. The accelerated lysosomal targeting of ApoD by oxidative stress is hindered in NPA fibroblasts, contributing to NPA lysosomes vulnerability. Exogenously added ApoD reduces NPA-prompted lysosomal permeabilization and alkalinization, reverts lipid peroxides accumulation, and significantly increases NPA cell survival. ApoD administered simultaneously to sphingomyelin overload results in complete rescue of cell survival. Our results reveal that ApoD protection of lysosomal integrity counteracts NPA pathology. ApoD supplementation could significantly delay not only the progression of NPA disease, but also of other LSDs through its beneficial effects in lysosomal functional maintenance.
Collapse
Affiliation(s)
- Raquel Pascua-Maestro
- Instituto de Biología y Genética Molecular, Universidad de Valladolid-CSIC, 47003 Valladolid, Spain
| | - Miriam Corraliza-Gomez
- Instituto de Biología y Genética Molecular, Universidad de Valladolid-CSIC, 47003 Valladolid, Spain
| | - Cristian Fadrique-Rojo
- Instituto de Biología y Genética Molecular, Universidad de Valladolid-CSIC, 47003 Valladolid, Spain
| | - Maria D Ledesma
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | | | - Diego Sanchez
- Instituto de Biología y Genética Molecular, Universidad de Valladolid-CSIC, 47003 Valladolid, Spain.
| | - Maria D Ganfornina
- Instituto de Biología y Genética Molecular, Universidad de Valladolid-CSIC, 47003 Valladolid, Spain.
| |
Collapse
|
42
|
Tazhitdinova R, Timoshenko AV. The Emerging Role of Galectins and O-GlcNAc Homeostasis in Processes of Cellular Differentiation. Cells 2020; 9:cells9081792. [PMID: 32731422 PMCID: PMC7465113 DOI: 10.3390/cells9081792] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023] Open
Abstract
Galectins are a family of soluble β-galactoside-binding proteins with diverse glycan-dependent and glycan-independent functions outside and inside the cell. Human cells express twelve out of sixteen recognized mammalian galectin genes and their expression profiles are very different between cell types and tissues. In this review, we summarize the current knowledge on the changes in the expression of individual galectins at mRNA and protein levels in different types of differentiating cells and the effects of recombinant galectins on cellular differentiation. A new model of galectin regulation is proposed considering the change in O-GlcNAc homeostasis between progenitor/stem cells and mature differentiated cells. The recognition of galectins as regulatory factors controlling cell differentiation and self-renewal is essential for developmental and cancer biology to develop innovative strategies for prevention and targeted treatment of proliferative diseases, tissue regeneration, and stem-cell therapy.
Collapse
|
43
|
Galectin-3 Stimulates Tyro3 Receptor Tyrosine Kinase and Erk Signalling, Cell Survival and Migration in Human Cancer Cells. Biomolecules 2020; 10:biom10071035. [PMID: 32664510 PMCID: PMC7407973 DOI: 10.3390/biom10071035] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022] Open
Abstract
The TAM (Tyro3, Axl, MerTK) subfamily of receptor tyrosine kinases (RTKs) and their ligands, Gas6 and protein S (ProS1), are implicated in tumorigenesis and chemoresistance in various cancers. The β-galactoside binding protein galectin-3 (Gal-3), which is also implicated in oncogenesis, has previously been shown to be a ligand for MerTK. However, the selectivity of Gal-3 for the other TAM receptors, and its TAM-mediated signalling and functional properties in cancer cells, remain to be explored. The present study was aimed at determining these, including through direct comparison of Gal-3 with the two canonical TAM ligands. Exogenous Gal-3 rapidly stimulated Tyro3 receptor phosphorylation to the same extent as the Tyro3 ligand ProS1, but not Axl, in the cultured human cancer cell lines SCC-25 (express both Tyro3 and Axl) and MGH-U3 (express Tyro3 only). Gal-3 also activated intracellular Erk and Akt kinases in both cell lines and furthermore protected cells from acute apoptosis induced by staurosporine but not from serum-starvation induced apoptosis. In addition, Gal-3 significantly stimulated cancer cell migration rate in the presence of the Axl blocker BGB324. Therefore, these results have shown Gal-3 to be a novel agonist for Tyro3 RTK, activating a Tyro3-Erk signalling axis, as well as Akt signalling, in cancer cells that promotes cell survival, cell cycle progression and cell migration. These data therefore reveal a novel mechanism of Tyro3 RTK activation through the action of Gal-3 that contrasts with those of the known TAM ligands Gas6 and ProS1.
Collapse
|
44
|
Puigdellívol M, Allendorf DH, Brown GC. Sialylation and Galectin-3 in Microglia-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2020; 14:162. [PMID: 32581723 PMCID: PMC7296093 DOI: 10.3389/fncel.2020.00162] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022] Open
Abstract
Microglia are brain macrophages that mediate neuroinflammation and contribute to and protect against neurodegeneration. The terminal sugar residue of all glycoproteins and glycolipids on the surface of mammalian cells is normally sialic acid, and addition of this negatively charged residue is known as “sialylation,” whereas removal by sialidases is known as “desialylation.” High sialylation of the neuronal cell surface inhibits microglial phagocytosis of such neurons, via: (i) activating sialic acid receptors (Siglecs) on microglia that inhibit phagocytosis and (ii) inhibiting binding of opsonins C1q, C3, and galectin-3. Microglial sialylation inhibits inflammatory activation of microglia via: (i) activating Siglec receptors CD22 and CD33 on microglia that inhibit phagocytosis and (ii) inhibiting Toll-like receptor 4 (TLR4), complement receptor 3 (CR3), and other microglial receptors. When activated, microglia release a sialidase activity that desialylates both microglia and neurons, activating the microglia and rendering the neurons susceptible to phagocytosis. Activated microglia also release galectin-3 (Gal-3), which: (i) further activates microglia via binding to TLR4 and TREM2, (ii) binds to desialylated neurons opsonizing them for phagocytosis via Mer tyrosine kinase, and (iii) promotes Aβ aggregation and toxicity in vivo. Gal-3 and desialylation may increase in a variety of brain pathologies. Thus, Gal-3 and sialidases are potential treatment targets to prevent neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - David H Allendorf
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
45
|
Galectins in the brain: advances in neuroinflammation, neuroprotection and therapeutic opportunities. Curr Opin Neurol 2020; 33:381-390. [DOI: 10.1097/wco.0000000000000812] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Srejovic I, Selakovic D, Jovicic N, Jakovljević V, Lukic ML, Rosic G. Galectin-3: Roles in Neurodevelopment, Neuroinflammation, and Behavior. Biomolecules 2020; 10:biom10050798. [PMID: 32455781 PMCID: PMC7277476 DOI: 10.3390/biom10050798] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
There is a plethora of evidence to suggest that Galectin-3 plays an important role in normal functions of mammalian cells, as well as in different pathogenic conditions. This review highlights recent data published by researchers, including our own team, on roles of Galectin-3 in the nervous system. Here, we discuss the roles of Galectin-3 in brain development, its roles in glial cells, as well as the interactions of glial cells with other neural and invading cells in pathological conditions. Galectin-3 plays an important role in the pathogenesis of neuroinflammatory and neurodegenerative disorders, such as multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. On the other hand, there is also evidence of the protective role of Galectin-3 due to its anti-apoptotic effect in target cells. Interestingly, genetic deletion of Galectin-3 affects behavioral patterns in maturing and adult mice. The results reviewed in this paper and recent development of highly specific inhibitors suggests that Galectin-3 may be an important therapeutic target in pathological conditions including the disorders of the central nervous system.
Collapse
Affiliation(s)
- Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia;
| | - Vladimir Jakovljević
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
- Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, 119146 Moscow, Russia
| | - Miodrag L. Lukic
- Department of Physiology—Molecular Medicine Unit, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Correspondence: (M.L.L.); (G.R.)
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
- Correspondence: (M.L.L.); (G.R.)
| |
Collapse
|
47
|
de Jong CGHM, Gabius HJ, Baron W. The emerging role of galectins in (re)myelination and its potential for developing new approaches to treat multiple sclerosis. Cell Mol Life Sci 2020; 77:1289-1317. [PMID: 31628495 PMCID: PMC7113233 DOI: 10.1007/s00018-019-03327-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system with unknown etiology. Currently approved disease-modifying treatment modalities are immunomodulatory or immunosuppressive. While the applied drugs reduce the frequency and severity of the attacks, their efficacy to regenerate myelin membranes and to halt disease progression is limited. To achieve such therapeutic aims, understanding biological mechanisms of remyelination and identifying factors that interfere with remyelination in MS can give respective directions. Such a perspective is given by the emerging functional profile of galectins. They form a family of tissue lectins, which are potent effectors in processes as diverse as adhesion, apoptosis, immune mediator release or migration. This review focuses on endogenous and exogenous roles of galectins in glial cells such as oligodendrocytes, astrocytes and microglia in the context of de- and (re)myelination and its dysregulation in MS. Evidence is arising for a cooperation among family members so that timed expression and/or secretion of galectins-1, -3 and -4 result in modifying developmental myelination, (neuro)inflammatory processes, de- and remyelination. Dissecting the mechanisms that underlie the distinct activities of galectins and identifying galectins as target or tool to modulate remyelination have the potential to contribute to the development of novel therapeutic strategies for MS.
Collapse
Affiliation(s)
- Charlotte G H M de Jong
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wia Baron
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
48
|
Extracellular Vesicles from Hyperammonemic Rats Induce Neuroinflammation and Motor Incoordination in Control Rats. Cells 2020; 9:cells9030572. [PMID: 32121257 PMCID: PMC7140428 DOI: 10.3390/cells9030572] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
Minimal hepatic encephalopathy is associated with changes in the peripheral immune system which are transferred to the brain, leading to neuroinflammation and thus to cognitive and motor impairment. Mechanisms by which changes in the immune system induce cerebral alterations remain unclear. Extracellular vesicles (EVs) seem to play a role in this process in certain pathologies. The aim of this work was to assess whether EVs play a role in the induction of neuroinflammation in cerebellum and motor incoordination by chronic hyperammonemia. We characterized the differences in protein cargo of EVs from plasma of hyperammonemic and control rats by proteomics and Western blot. We assessed whether injection of EVs from hyperammonemic to normal rats induces changes in neuroinflammation in cerebellum and motor incoordination similar to those exhibited by hyperammonemic rats. We found that hyperammonemia increases EVs amount and alters their protein cargo. Differentially expressed proteins are mainly associated with immune system processes. Injected EVs enter Purkinje neurons and microglia. Injection of EVs from hyperammonemic, but not from control rats, induces motor incoordination, which is mediated by neuroinflammation, microglia and astrocytes activation and increased IL-1β, TNFα, its receptor TNFR1, NF-κB in microglia, glutaminase I, and GAT3 in cerebellum. Plasma EVs from hyperammonemic rats carry molecules necessary and sufficient to trigger neuroinflammation in cerebellum and the mechanisms leading to motor incoordination.
Collapse
|
49
|
Yazar HO, Yazar T, Cihan M. A preliminary data: Evaluation of serum Galectin-3 levels in patients with Idiopathic Parkinson's Disease. J Clin Neurosci 2019; 70:164-168. [PMID: 31471077 DOI: 10.1016/j.jocn.2019.08.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Abstract
AIM In our study, we aimed to collect data for the hypothesis that Galectin-3 might be used as a new prognostic and therapeutic biomarker in Idiopathic Parkinson's Disease (IPD). METHOD In this prospective and cross-sectional study, the Unified Parkinson's Disease Rating Scale (UPDRS) and Modified Hoehn and Yahr (H&Y) scales were applied to each patient diagnosed as IPD according to the UK Brain Bank diagnostic criteria. The control group consisted of healthy individuals with the same age, gender, and body mass index characteristics as the patients meeting the exclusion criteria. RESULTS A total of 111 cases were included in the study, 48 were IPD, and 63 were healthy controls. There were no statistically significant differences between the IPD and control groups in terms of demographic, anthropometric, and blood parameters (p > 0.05). Serum galectin-3 levels were significantly higher in IPD than the control group (p < 0.001). Serum galectin-3 levels, UPDRS scores, and duration of disease were significantly higher in patients with IPD in parallel with the progression of the disease (p < 0.001; 0.001; 0.009). No significant relationship was detected between the stage of the disease and other parameters (p < 0.05). CONCLUSION Our study supports the hypothesis that serum galectin-3 level might be associated with IPD. Our data suggest that serum galectin-3 levels might be an accessible biomarker for the detection and prevention of chronic, progressive diseases such as IPH.
Collapse
Affiliation(s)
- Hülya Olgun Yazar
- Ordu University Training and Research Hospital, Clinic of Neurology, Turkey.
| | - Tamer Yazar
- Ordu State Hospital, Clinic of Neurology, Turkey
| | - Murat Cihan
- Ordu University Training and Research Hospital, Clinical Biochemist, Turkey
| |
Collapse
|
50
|
Wang Q, Gao R, Wang M, Chen Q, Xiao M, Li Z, Wang L, Chen C. Spatiotemporal expression patterns of Galectin-3 in perinatal rat hypoxic-ischemic brain injury model. Neurosci Lett 2019; 711:134439. [PMID: 31425825 DOI: 10.1016/j.neulet.2019.134439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/03/2019] [Accepted: 08/15/2019] [Indexed: 12/28/2022]
Abstract
In this research, we intended to evaluate the expression pattern, distribution and sources of Galectin-3 (Gal-3) in perinatal hypoxic-ischemic brain injury rat model. Postnatal day 3 Sprague-Dawley rat pups were subjected to right carotid artery ligation followed by 2.5 h of hypoxia (6% oxygen). Expression and distribution of Gal-3 were evaluated by western blotting and immunofluorescence. Sources of Gal-3 were evaluated by double staining with neuronic, oligodendrocytic, astrocytic, microglial and endotheliocytic markers. Our results indicated Gal-3 significantly upregulated from 12 h and maintained an increasing tendency within 72 h post injury. Although the relative expression of Gal-3 decreased after 72 h, we detected significant differences until 14d. We found Gal-3 started to distribute in cortex and thalamus area and maintained an increasing tendency. Gal-3 could be detected in cortex, thalamus, corpus callosum and hippocampus area at 72 h post injury. After that, expression of Gal-3 in cortex and thalamus area downregulated, the expression in corpus callosum and hippocampus area vanished. We found astrocyte, microglia, neuron and endotheliocyte were sources of Gal-3 in cortex area; astrocyte, microglia and endotheliocyte were sources of Gal-3 in thalamus area; oligodendrocyte precursor cell and endotheliocyte were sources of Gal-3 in corpus callosum; neuron, microglia and endotheliocyte were sources of Gal-3 in hippocampus. In conclusion, we demonstrated spatiotemporal expression patterns of Galectin-3 post perinatal hypoxic-ischemic brain injury in this research.
Collapse
Affiliation(s)
- Qian Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Ruiwei Gao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Minjie Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Qiufan Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Mili Xiao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Zhihua Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Laishuan Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China.
| |
Collapse
|