1
|
Min E, Ko MY, Kim M, Park H, Kim Y, Kim KK, Lee BS, Hyun SA, Ka M. Perfluorooctanoic acid (PFOA) activates astrogliosis-associated neuroinflammation through ER stress-autophagy axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 979:179546. [PMID: 40288163 DOI: 10.1016/j.scitotenv.2025.179546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Perfluorooctanoic acid (PFOA), a perfluoroalkyl acid, induces neuroinflammation. However, present understanding regarding its fundamental role in neuroinflammation remains limited. Therefore, in this study, we aimed to clarify the potential association between PFOA and astrocyte activation via the modulation of the endoplasmic reticulum (ER) stress-autophagy axis. The results obtained revealed that PFOA activated astroglia in A-172 astrocytoma cells and primary astrocytes by upregulating the expression levels of autophagy-related proteins (ATG5, BECN1, SQSTM1, and MAP1LC3B-II). It also activated autophagy in A-172 astrocytoma cells and primary astrocytes via the upstream activation of ER stress-related proteins, such as ATF4, GRP78, and CHOP. Further, the pharmacological inhibition of ER stress as well as autophagy prevented PFOA-induced activation of astrogliosis in PFOA-treated A-172 cells and primary astrocytes. We also observed that PFOA-mediated activation of GFAP upregulated the transcription of pro-inflammatory cytokines, such as IL-1β, TNF-α, and IL-6. These findings confirmed the existence of a relationship between ER stress-induced autophagy and astrogliosis in PFOA-treated astrocytes, suggesting that targeting the ER stress-autophagy axis may be a potential therapeutic strategy for reducing PFOA-induced neuroinflammation.
Collapse
Affiliation(s)
- Euijun Min
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moon Yi Ko
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Minjeong Kim
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Heejin Park
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Younhee Kim
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kee K Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Byoung-Seok Lee
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Sung-Ae Hyun
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Minhan Ka
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34114, Republic of Korea.
| |
Collapse
|
2
|
Malone K, LaCasse E, Beug ST. Cell death in glioblastoma and the central nervous system. Cell Oncol (Dordr) 2025; 48:313-349. [PMID: 39503973 PMCID: PMC11997006 DOI: 10.1007/s13402-024-01007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma is the commonest and deadliest primary brain tumor. Glioblastoma is characterized by significant intra- and inter-tumoral heterogeneity, resistance to treatment and dismal prognoses despite decades of research in understanding its biological underpinnings. Encompassed within this heterogeneity and therapy resistance are severely dysregulated programmed cell death pathways. Glioblastomas recapitulate many neurodevelopmental and neural injury responses; in addition, glioblastoma cells are composed of multiple different transformed versions of CNS cell types. To obtain a greater understanding of the features underlying cell death regulation in glioblastoma, it is important to understand the control of cell death within the healthy CNS during homeostatic and neurodegenerative conditions. Herein, we review apoptotic control within neural stem cells, astrocytes, oligodendrocytes and neurons and compare them to glioblastoma apoptotic control. Specific focus is paid to the Inhibitor of Apoptosis proteins, which play key roles in neuroinflammation, CNS cell survival and gliomagenesis. This review will help in understanding glioblastoma as a transformed version of a heterogeneous organ composed of multiple varied cell types performing different functions and possessing different means of apoptotic control. Further, this review will help in developing more glioblastoma-specific treatment approaches and will better inform treatments looking at more direct brain delivery of therapeutic agents.
Collapse
Affiliation(s)
- Kyle Malone
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Eric LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
3
|
Musotto R, Wanderlingh U, Pioggia G. Ca 2+ waves in astrocytes: computational modeling and experimental data. Front Cell Neurosci 2025; 19:1536096. [PMID: 40226297 PMCID: PMC11985530 DOI: 10.3389/fncel.2025.1536096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
This paper examines different computational models for Calcium wave propagation in astrocytes. Through a comparative analysis of models by Goldbeter, De Young-Keizer, Atri, Li-Rinzel, and De Pittà and of experimental data, the study highlights the model contributions for the understanding of Calcium dynamics. Tracing the evolution from simple to complex models, this work emphasizes the importance of integrating experimental data in order to further refine these models. The results allow to improve our understanding of the physiological functions of astrocytes, suggesting the importance of more accurate astrocyte models.
Collapse
Affiliation(s)
- Rosa Musotto
- National Research Council, IRIB-CNR, Institute for Biomedical Research and Innovation, Messina, Italy
| | - Ulderico Wanderlingh
- Department of Mathematical and Computer Sciences, Physical Sciences and Earth Sciences, University of Messina, Messina, Italy
| | - Giovanni Pioggia
- National Research Council, IRIB-CNR, Institute for Biomedical Research and Innovation, Messina, Italy
| |
Collapse
|
4
|
Gilloteaux J, Charlier C, Suain V, Nicaise C. Astrocyte alterations during Osmotic Demyelination Syndrome: intermediate filaments, aggresomes, proteasomes, and glycogen storages. Ultrastruct Pathol 2025; 49:170-215. [PMID: 40062739 DOI: 10.1080/01913123.2025.2468700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION A murine model mimicking the human osmotic demyelination syndrome (ODS) revealed with histology demyelinated alterations in the relay posterolateral (VPL) and ventral posteromedial (VPM) thalamic nuclei 12 h and 48 h after chronic hyponatremia due to a fast reinstatement of osmolality. Abnormal expression astrocyte markers ALDHL1 and GFAP with immunohistochemistry in these ODS altered zones, prompted aims to verify in both protoplasmic and fibrillar astrocytes with ultrastructure those changes and other associated subcellular modifications. METHOD This ODS investigation included four groups of mice: Sham (NN; n = 13), hyponatremic (HN; n = 11), those sacrificed 12 h after a fast restoration of normal natremia (ODS12h; n = 6), and mice sacrificed 48 h afterward, or ODS48 h (n = 9). Out of those four groups of mice, with LM and ultrastructure microscopy, the thalamic zones included NN (n = 2), HN (n = 2), ODS12h (n = 3) and ODS48h (n = 3) samples. There, comparisons between astrocytes included organelles, GFAP, and glycogen content changes. RESULTS Thalamic ODS epicenter damages comprised both protoplasmic (PA) and fibrillar (FA) astrocyte necroses along with those of neuropil destructions and neuron Wallerian demyelinated injuries surrounded by a centrifugal region gradient revealing worse to mild destructions. Ultrastructure aspects of resilient HN and ODS12h PAs disclosed altered mitochondria and accumulations of beta- to alpha-glycogen granules that became eventually captured into phagophores as glycophagosomes in ODS48h. HN and ODS12h time lapse FAs accumulated ribonucleoproteins, cytoskeletal aggresomes, and proteasomes but distant and resilient ODS48h FAs maintained GFAP fibrils along with typical mitochondria and dispersed β-glycogen, including in their neuropil surroundings. Thus, ODS triggered astrocyte injuries that involved both post-transcriptional and post-translational modifications such that astrocytes were unable to use glycogen and metabolites due to their own mitochondria defects while accumulated stalled ribonucleoproteins, cytoskeletal aggresomes were associated with proteasomes and GFAP ablation. Resilient but distant astrocytes revealed restitution of amphibolism where typical carbohydrate storages were revealed along with GFAP, as tripartite extensions supply for restored nerve axon initial segments, neural Ranvier's junctions, and oligodendrocyte -neuron junctional contacts. CONCLUSION ODS caused astrocyte damage associated with adjacent neuropil destruction that included a regional demyelination caused by a loss of dispatched energetic and metabolic exchanges within the injured region, bearing proportional and collateral centrifugal injuries, which involved reactive repairs time after rebalanced osmolarity.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- Department of Medicine, Laboratory of Neurodegeneration and Regeneration URPHyM, NARILIS, University of Namur, Namur, Belgium
- Department of Anatomical Sciences, St George's University School of Medicine, KB Taylor Global Scholar's Program at the Northumbria University, Newcastle upon Tyne, UK
| | - Corry Charlier
- Electron Microscopy Platform, MORPH-IM, Université de Namur, Bruxelles, Belgium
| | - Valérie Suain
- CMMI - The Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Charles Nicaise
- Department of Medicine, Laboratory of Neurodegeneration and Regeneration URPHyM, NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
5
|
Osterman C, Hamlin D, Suter CM, Affleck AJ, Gloss BS, Turner CP, Faull RLM, Stein TD, McKee A, Buckland ME, Curtis MA, Murray HC. Perivascular glial reactivity is a feature of phosphorylated tau lesions in chronic traumatic encephalopathy. Acta Neuropathol 2025; 149:16. [PMID: 39921702 PMCID: PMC11807024 DOI: 10.1007/s00401-025-02854-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Chronic traumatic encephalopathy (CTE), a neurodegenerative disease associated with repetitive head injuries, is characterised by perivascular hyperphosphorylated tau (p-tau) accumulations within the depths of cortical sulci. Although the majority of CTE literature focuses on p-tau pathology, other pathological features such as glial reactivity, vascular damage, and axonal damage are relatively unexplored. In this study, we aimed to characterise these other pathological features, specifically in CTE p-tau lesion areas, to better understand the microenvironment surrounding the lesion. We utilised multiplex immunohistochemistry to investigate the distribution of 32 different markers of cytoarchitecture and pathology that are relevant to both traumatic brain injury and neurodegeneration. We qualitatively assessed the multiplex images and measured the percentage area of labelling for each marker in the lesion and non-lesion areas of CTE cases. We identified perivascular glial reactivity as a prominent feature of CTE p-tau lesions, largely driven by increases in astrocyte reactivity compared to non-lesion areas. Furthermore, we identified astrocytes labelled for both NAD(P)H quinone dehydrogenase 1 (NQO1) and L-ferritin, indicating that lesion-associated glial reactivity may be a compensatory response to iron-induced oxidative stress. Our findings demonstrate that perivascular inflammation is a consistent feature of the CTE pathognomonic lesion and may contribute to the pathogenesis of brain injury-related neurodegeneration.
Collapse
Affiliation(s)
- Chelsie Osterman
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Danica Hamlin
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Catherine M Suter
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Andrew J Affleck
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Brian S Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Clinton P Turner
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
- Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, 2 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Richard L M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Thor D Stein
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Ann McKee
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Michael E Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, 94 Mallet St, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, 85 Park Road, Grafton, 1023, Auckland, New Zealand.
| |
Collapse
|
6
|
Cheng Q, Liu Y, Yang Z, Zhang M, Liu T, Niu Y, Liu W, Huang L, Feng Y, Zhang X, Luo X, Ning Q, Chen T. Evaluation of Plasma Neurodegenerative Biomarkers for Diagnosing Minimal Hepatic Encephalopathy and Predicting Overt Hepatic Encephalopathy in Chinese Patients with Hepatic Cirrhosis. J Clin Transl Hepatol 2025; 13:35-46. [PMID: 39801785 PMCID: PMC11712093 DOI: 10.14218/jcth.2024.00413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Background and Aims The performance of neurodegenerative biomarkers-neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), tau, and ubiquitin carboxy-terminal hydrolase L1 (UCHL1)-in diagnosing minimal hepatic encephalopathy (MHE) has not been systematically evaluated, simultaneously, nor have their associations with the development of overt hepatic encephalopathy (OHE). This study aimed to evaluate the performance of plasma NfL, GFAP, tau, and UCHL1 in diagnosing MHE and predicting the development of OHE in Chinese patients with hepatic cirrhosis. Methods In this prospective study, 124 patients with hepatic cirrhosis were recruited. The Psychometric Hepatic Encephalopathy Score was used to diagnose MHE, and OHE development was observed during a 30-day follow-up period. Plasma levels of NfL, GFAP, tau, and UCHL1 were measured using the highly sensitive single-molecule array when MHE was diagnosed. Additionally, serum interleukin-6 (IL-6) levels and the model for end-stage liver disease (MELD) and MELD-Na scores were also measured. Results MHE was diagnosed in 57 (46.0%) patients. Patients with MHE had significantly higher plasma levels of NfL and GFAP (34.2 vs. 22.4 pg/mL and 173 vs. 97.6 pg/mL, respectively; both p < 0.001) and lower tau levels (8.4 vs. 11.6 pg/mL, p = 0.048) compared to those without MHE. Plasma NfL (odds ratios = 1.027, 95% confidence interval [CI]: 1.006-1.048; p = 0.013) and serum ammonia levels (odds ratios = 1.021, 95% CI: 1.006-1.036; p = 0.007) were independently associated with MHE occurrence. A combination of NfL, GFAP, tau, and UCHL1 was effective in diagnosing MHE in all cirrhotic patients (area under the receiver operating characteristic curve [hereinafter referred to as AUROC]: 0.748, 95% CI: 0.662-0.821), with an accuracy, sensitivity, and specificity of 71.0%, 71.9%, and 71.6%, respectively. In patients without previous OHE, the combination had an AUROC of 0.764 (95% CI: 0.673-0.840), with an accuracy, sensitivity, and specificity of 72.5%, 71.7%, and 73.0%, respectively. Furthermore, GFAP (hazard ratio (HR) = 1.003, 95% CI: 1.000-1.005; p = 0.044), IL-6 (HR = 1.003, 95% CI: 1.001-1.004; p < 0.001), and MELD score (HR = 1.139, 95% CI: 1.072-1.210; p < 0.001)-but not NfL, tau, and UCHL1-were identified as risk factors for 30-day OHE development. Conclusions The combination of plasma levels of NfL, GFAP, tau, and UCHL1 performs well in diagnosing MHE. Additionally, MELD score, IL-6, and GFAP appear to be significant predictors of OHE development in patients with hepatic cirrhosis.
Collapse
Affiliation(s)
- Qiuyu Cheng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunhui Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongyuan Yang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng Zhang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuxin Niu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Liu
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lanyue Huang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuzhao Feng
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyun Zhang
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Ning
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Chen
- Department of Infectious Diseases, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
7
|
Gogishvili D, Honey MIJ, Verberk IMW, Vermunt L, Hol EM, Teunissen CE, Abeln S. The GFAP proteoform puzzle: How to advance GFAP as a fluid biomarker in neurological diseases. J Neurochem 2025; 169:e16226. [PMID: 39289040 DOI: 10.1111/jnc.16226] [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: 06/05/2024] [Revised: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Glial fibrillary acidic protein (GFAP) is a well-established biomarker of reactive astrogliosis in the central nervous system because of its elevated levels following brain injury and various neurological disorders. The advent of ultra-sensitive methods for measuring low-abundant proteins has significantly enhanced our understanding of GFAP levels in the serum or plasma of patients with diverse neurological diseases. Clinical studies have demonstrated that GFAP holds promise both as a diagnostic and prognostic biomarker, including but not limited to individuals with Alzheimer's disease. GFAP exhibits diverse forms and structures, herein referred to as its proteoform complexity, encompassing conformational dynamics, isoforms and post-translational modifications (PTMs). In this review, we explore how the proteoform complexity of GFAP influences its detection, which may affect the differential diagnostic performance of GFAP in different biological fluids and can provide valuable insights into underlying biological processes. Additionally, proteoforms are often disease-specific, and our review provides suggestions and highlights areas to focus on for the development of new assays for measuring GFAP, including isoforms, PTMs, discharge mechanisms, breakdown products, higher-order species and interacting partners. By addressing the knowledge gaps highlighted in this review, we aim to support the clinical translation and interpretation of GFAP in both CSF and blood and the development of reliable, reproducible and specific prognostic and diagnostic tests. To enhance disease pathology comprehension and optimise GFAP as a biomarker, a thorough understanding of detected proteoforms in biofluids is essential.
Collapse
Affiliation(s)
- Dea Gogishvili
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Madison I J Honey
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Sanne Abeln
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
8
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
9
|
Abdelaziz MA, Chen WH, Chang YW, Mindaye SA, Chen CC. Exploring the role of spinal astrocytes in the onset of hyperalgesic priming signals in acid-induced chronic muscle pain. PNAS NEXUS 2024; 3:pgae362. [PMID: 39228816 PMCID: PMC11370897 DOI: 10.1093/pnasnexus/pgae362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024]
Abstract
Hyperalgesic priming, a form of pain plasticity initiated by initial injury, leads to heightened sensitivity to subsequent noxious stimuli, contributing to chronic pain development in animals. While astrocytes play active roles in modulating synaptic transmission in various pain models, their specific involvement in hyperalgesic priming remains elusive. Here, we show that spinal astrocytes are essential for hyperalgesic priming formation in a mouse model of acid-induced muscle pain. We observed spinal astrocyte activation 4 h after initial acid injection, and inhibition of this activation prevented chronic pain development upon subsequent acid injection. Chemogenetic activation of spinal astrocytes mimicked the first acid-induced hyperalgesic priming. We also demonstrated that spinal phosphorylated extracellular regulated kinase (pERK)-positive neurons were mainly vesicular glutamate transporter-2 positive (Vglut2+) neurons after the first acid injection, and inhibition of spinal pERK prevented astrocyte activation. Furthermore, pharmacological inhibition of astrocytic glutamate transporters glutamate transporter-1 and glutamate-aspartate transporter abolished the hyperalgesic priming. Collectively, our results suggest that pERK activation in Vglut2+ neurons activate astrocytes through astrocytic glutamate transporters. This process eventually establishes hyperalgesic priming through spinal D-serine. We conclude that spinal astrocytes play a crucial role in the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Mohamed Abbas Abdelaziz
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Zoology Department, Faculty of Science, Al-Azhar University Assiut Branch, Assiut 71524, Egypt
| | - Wei-Hsin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Selomon Assefa Mindaye
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
10
|
Gogishvili D, Illes-Toth E, Harris MJ, Hopley C, Teunissen CE, Abeln S. Structural flexibility and heterogeneity of recombinant human glial fibrillary acidic protein (GFAP). Proteins 2024; 92:649-664. [PMID: 38149328 DOI: 10.1002/prot.26656] [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: 10/04/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023]
Abstract
Glial fibrillary acidic protein (GFAP) is a promising biomarker for brain and spinal cord disorders. Recent studies have highlighted the differences in the reliability of GFAP measurements in different biological matrices. The reason for these discrepancies is poorly understood as our knowledge of the protein's 3-dimensional conformation, proteoforms, and aggregation remains limited. Here, we investigate the structural properties of GFAP under different conditions. For this, we characterized recombinant GFAP proteins from various suppliers and applied hydrogen-deuterium exchange mass spectrometry (HDX-MS) to provide a snapshot of the conformational dynamics of GFAP in artificial cerebrospinal fluid (aCSF) compared to the phosphate buffer. Our findings indicate that recombinant GFAP exists in various conformational species. Furthermore, we show that GFAP dimers remained intact under denaturing conditions. HDX-MS experiments show an overall decrease in H-bonding and an increase in solvent accessibility of GFAP in aCSF compared to the phosphate buffer, with clear indications of mixed EX2 and EX1 kinetics. To understand possible structural interface regions and the evolutionary conservation profiles, we combined HDX-MS results with the predicted GFAP-dimer structure by AlphaFold-Multimer. We found that deprotected regions with high structural flexibility in aCSF overlap with predicted conserved dimeric 1B and 2B domain interfaces. Structural property predictions combined with the HDX data show an overall deprotection and signatures of aggregation in aCSF. We anticipate that the outcomes of this research will contribute to a deeper understanding of the structural flexibility of GFAP and ultimately shed light on its behavior in different biological matrices.
Collapse
Affiliation(s)
- Dea Gogishvili
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Eva Illes-Toth
- National Measurement Laboratory at Laboratory of the Government Chemist (LGC), Teddington, UK
| | - Matthew J Harris
- National Measurement Laboratory at Laboratory of the Government Chemist (LGC), Teddington, UK
| | - Christopher Hopley
- National Measurement Laboratory at Laboratory of the Government Chemist (LGC), Teddington, UK
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sanne Abeln
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
11
|
Parsi S, Zhu C, Motlagh NJ, Kim D, Küllenberg EG, Kim HH, Gillani RL, Chen JW. Basic Science of Neuroinflammation and Involvement of the Inflammatory Response in Disorders of the Nervous System. Magn Reson Imaging Clin N Am 2024; 32:375-384. [PMID: 38555147 PMCID: PMC10987041 DOI: 10.1016/j.mric.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Neuroinflammation is a key immune response observed in many neurologic diseases. Although an appropriate immune response can be beneficial, aberrant activation of this response recruits excessive proinflammatory cells to cause damage. Because the central nervous system is separated from the periphery by the blood-brain barrier (BBB) that creates an immune-privileged site, it has its own unique immune cells and immune response. Moreover, neuroinflammation can compromise the BBB causing an influx of peripheral immune cells and factors. Recent advances have brought a deeper understanding of neuroinflammation that can be leveraged to develop more potent therapies and improve patient selection.
Collapse
Affiliation(s)
- Sepideh Parsi
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cindy Zhu
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Negin Jalali Motlagh
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daeki Kim
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Enrico G Küllenberg
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyung-Hwan Kim
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca L Gillani
- Department of Neurology, Neuroimmunology and Neuro-Infectious Diseases Division, Massachusetts Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John W Chen
- Institute for Innovation in Imaging, Neurovascular Research Unit, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Hamani C, Davidson B, Lipsman N, Abrahao A, Nestor SM, Rabin JS, Giacobbe P, Pagano RL, Campos ACP. Insertional effect following electrode implantation: an underreported but important phenomenon. Brain Commun 2024; 6:fcae093. [PMID: 38707711 PMCID: PMC11069120 DOI: 10.1093/braincomms/fcae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/08/2023] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Deep brain stimulation has revolutionized the treatment of movement disorders and is gaining momentum in the treatment of several other neuropsychiatric disorders. In almost all applications of this therapy, the insertion of electrodes into the target has been shown to induce some degree of clinical improvement prior to stimulation onset. Disregarding this phenomenon, commonly referred to as 'insertional effect', can lead to biased results in clinical trials, as patients receiving sham stimulation may still experience some degree of symptom amelioration. Similar to the clinical scenario, an improvement in behavioural performance following electrode implantation has also been reported in preclinical models. From a neurohistopathologic perspective, the insertion of electrodes into the brain causes an initial trauma and inflammatory response, the activation of astrocytes, a focal release of gliotransmitters, the hyperexcitability of neurons in the vicinity of the implants, as well as neuroplastic and circuitry changes at a distance from the target. Taken together, it would appear that electrode insertion is not an inert process, but rather triggers a cascade of biological processes, and, as such, should be considered alongside the active delivery of stimulation as an active part of the deep brain stimulation therapy.
Collapse
Affiliation(s)
- Clement Hamani
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Benjamin Davidson
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Nir Lipsman
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Agessandro Abrahao
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Sean M Nestor
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Jennifer S Rabin
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto M5G 1V7, Canada
| | - Peter Giacobbe
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Rosana L Pagano
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo, SP CEP 01308-060, Brazil
| | - Ana Carolina P Campos
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo, SP CEP 01308-060, Brazil
| |
Collapse
|
13
|
Turniak-Kusy M, Studzian M, Szpakowski P, Kuchta P, Smietanka K, Mattern C, Pulaski L, Bielecki B. Testosterone Inhibits Secretion of the Pro-Inflammatory Chemokine CXCL1 from Astrocytes. Curr Issues Mol Biol 2024; 46:2105-2118. [PMID: 38534751 DOI: 10.3390/cimb46030135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
Astrocytes play an important role in the regulation of the inflammatory response in the CNS, e.g., in demyelinating diseases. Since the chemokine CXCL1 is known to be secreted by astrocytes and to have a pro-inflammatory effect on immune cells in the CNS, we verified the effect of testosterone on its secretion in vitro (in the astrocytic cell line DI TNC1). Testosterone reduced the increase in CXCL1 production caused by the pro-inflammatory agent lysophosphatidylcholine and restored the basal production level of CXCL1. The androgen receptor (present and functional in the studied cell line) was strongly suggested to mediate this effect-its non-steroid ligand flutamide exerted an agonist-like effect, mimicking the activity of testosterone itself on CXCL1 secretion. This novel mechanism has important implications for the known immunomodulatory effect of testosterone and potentially other androgenic hormones. It provides a potential explanation on the molecular level and shows that astrocytes are important players in inflammatory homeostasis in the CNS and its hormonal regulation. Therefore, it suggests new directions for the development of the therapeutic intervention.
Collapse
Affiliation(s)
| | - Maciej Studzian
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Piotr Kuchta
- Faculty of Medicine, Medical University of Lodz, 90-419 Lodz, Poland
| | - Kaja Smietanka
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Claudia Mattern
- Oceanographic Center, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- M&P Pharma AG, 6376 Emmetten, Switzerland
| | - Lukasz Pulaski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Bartosz Bielecki
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
14
|
Arya S, Bahuguna D, Bajad G, Loharkar S, Devangan P, Khatri DK, Singh SB, Madan J. Colloidal therapeutics in the management of traumatic brain injury: Portray of biomarkers and drug-targets, preclinical and clinical pieces of evidence and future prospects. Colloids Surf B Biointerfaces 2023; 230:113509. [PMID: 37595379 DOI: 10.1016/j.colsurfb.2023.113509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/20/2023]
Abstract
Complexity associated with the aberrant physiology of traumatic brain injury (TBI) makes its therapeutic targeting vulnerable. The underlying mechanisms of pathophysiology of TBI are yet to be completely illustrated. Primary injury in TBI is associated with contusions and axonal shearing whereas excitotoxicity, mitochondrial dysfunction, free radicals generation, and neuroinflammation are considered under secondary injury. MicroRNAs, proinflammatory cytokines, and Glial fibrillary acidic protein (GFAP) recently emerged as biomarkers in TBI. In addition, several approved therapeutic entities have been explored to target existing and newly identified drug-targets in TBI. However, drug delivery in TBI is hampered due to disruption of blood-brain barrier (BBB) in secondary TBI, as well as inadequate drug-targeting and retention effect. Colloidal therapeutics appeared helpful in providing enhanced drug availability to the brain owing to definite targeting strategies. Moreover, immense efforts have been put together to achieve increased bioavailability of therapeutics to TBI by devising effective targeting strategies. The potential of colloidal therapeutics to efficiently deliver drugs at the site of injury and down-regulate the mediators of TBI are serving as novel policies in the management of TBI. Therefore, in present manuscript, we have illuminated a myriad of molecular-targets currently identified and recognized in TBI. Moreover, particular emphasis is given to frame armamentarium of repurpose drugs which could be utilized to block molecular targets in TBI in addition to drug delivery barriers. The critical role of colloidal therapeutics such as liposomes, nanoparticles, dendrimers, and exosomes in drug delivery to TBI through invasive and non-invasive routes has also been highlighted.
Collapse
Affiliation(s)
- Shristi Arya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Deepankar Bahuguna
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Gopal Bajad
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Soham Loharkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Pawan Devangan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
15
|
Felis-Giemza A, Massalska M, Roszkowski L, Romanowska-Próchnicka K, Ciechomska M. Potential Mechanism of Fatigue Induction and Its Management by JAK Inhibitors in Inflammatory Rheumatic Diseases. J Inflamm Res 2023; 16:3949-3965. [PMID: 37706062 PMCID: PMC10497048 DOI: 10.2147/jir.s414739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/27/2023] [Indexed: 09/15/2023] Open
Abstract
It is well known that fatigue is a highly disabling symptom commonly observed in inflammatory rheumatic diseases (IRDs). Fatigue is strongly associated with a poor quality of life and seems to be an independent predictor of job loss and disability in patients with different rheumatic diseases. Although the pathogenesis of fatigue remains unclear, indirect data suggest the cooperation of the immune system, the central and autonomic nervous system, and the neuroendocrine system in the induction and sustainment of fatigue in chronic diseases. Fatigue does not correspond with disease activity and its mechanism in IRDs. It is suggested that it may change over time and vary between individuals. Abnormal production of pro-inflammatory cytokines such as interleukin-6 (IL-6), interferons (IFNs), granulocyte-macrophage colony-stimulating factor (GM-CSF), TNF, IL-15, IL-17 play a role in both IRDs and subsequent fatigue development. Some of these cytokines such as IL-6, IFNs, GM-CSF, and common gamma-chain cytokines (IL-15, IL-2, and IL-7) activate the Janus Kinases (JAKs) family of intracellular tyrosine kinases. Therapy blocking JAKs (JAK inhibitors - JAKi) has been recently proven to be an effective approach for IRDs treatment, more efficient in pain reduction than anti-TNF. Therefore, the administration of JAKi to IRDs patients experiencing fatigue may find rational implications as a therapeutic modulator not only of disease inflammatory symptoms but also fatigue with its components like pain and neuropsychiatric features as well. In this review, we demonstrate the latest information on the mechanisms of fatigue in rheumatic diseases and the potential effect of JAKi on fatigue reduction.
Collapse
Affiliation(s)
- Anna Felis-Giemza
- Biologic Therapy Center, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, Poland
| | - Magdalena Massalska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, Poland
| | - Leszek Roszkowski
- Department of Outpatient Clinics, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, Poland
| | - Katarzyna Romanowska-Próchnicka
- Department of Biophysics, Physiology and Pathophysiology, Faculty of Health Sciences, Warsaw Medical University, Warsaw, Poland
| | - Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology, and Rehabilitation (NIGRiR), Warsaw, Poland
| |
Collapse
|
16
|
Le Berre C, Naveilhan P, Rolli-Derkinderen M. Enteric glia at center stage of inflammatory bowel disease. Neurosci Lett 2023; 809:137315. [PMID: 37257681 DOI: 10.1016/j.neulet.2023.137315] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
Although our understanding of the pathophysiology of inflammatory bowel disease (IBD) is increasing, the expanding body of knowledge does not simplify the equation but rather reveals diverse, interconnected, and complex mechanisms in IBD. In addition to immune overactivation, defects in intestinal epithelial barrier (IEB) functioning, dysbiosis, and structural and functional abnormalities of the enteric nervous system are emerging as new elements contributing to the development of IBD. In addition to molecular changes in IBD, enteric glia from patients with Crohn's disease (CD) exhibits the inability to strengthen the IEB; these defects are not observed in patients with ulcerative colitis. In addition, there is a growing body of work describing that enteric glia interacts with not only enterocytes and enteric neurons but also other local cellular neighbours. Thus, because of their functions as connectors and regulators of immune cells, IEB, and microbiota, enteric glia could be the keystone of digestive homeostasis that is lacking in patients with CD.
Collapse
Affiliation(s)
- Catherine Le Berre
- Hépato-Gastro-Entérologie et Assistance Nutritionnelle, Inserm CIC 1413, Institut des Maladies de l'Appareil Digestif (IMAD), CHU Nantes, 1 place Alexis Ricordeau, F-44000 Nantes, France; Nantes Université, CHU Nantes, INSERM, The Enteric Nervous System in Gut and Brain Disorders, IMAD, 1 rue Gaston Veil, 44035 Nantes Cedex 1, F-44000 Nantes, France
| | - Philippe Naveilhan
- Nantes Université, CHU Nantes, INSERM, The Enteric Nervous System in Gut and Brain Disorders, IMAD, 1 rue Gaston Veil, 44035 Nantes Cedex 1, F-44000 Nantes, France
| | - Malvyne Rolli-Derkinderen
- Nantes Université, CHU Nantes, INSERM, The Enteric Nervous System in Gut and Brain Disorders, IMAD, 1 rue Gaston Veil, 44035 Nantes Cedex 1, F-44000 Nantes, France.
| |
Collapse
|
17
|
Wang LY, Wang XP, Lv JM, Shan YD, Jia SY, Yu ZF, Miao HT, Xin Y, Zhang DX, Zhang LM. NLRP3-GABA signaling pathway contributes to the pathogenesis of impulsive-like behaviors and cognitive deficits in aged mice. J Neuroinflammation 2023; 20:162. [PMID: 37434240 DOI: 10.1186/s12974-023-02845-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND), such as delirium and cognitive impairment, are commonly encountered complications in aged patients. The inhibitory neurotransmitter γ-aminobutyric acid (GABA) is aberrantly synthesized from reactive astrocytes following inflammatory stimulation and is implicated in the pathophysiology of neurodegenerative diseases. Additionally, the activation of NOD-like receptor protein 3 (NLRP3) inflammasome is involved in PND. Herein, we aimed to investigate whether the NLRP3-GABA signaling pathway contributes to the pathogenesis of aging mice's PND. METHODS 24-month-old C57BL/6 and astrocyte-specific NLRP3 knockout male mice were used to establish a PND model via tibial fracture surgery. The monoamine oxidase-B (MAOB) inhibitor selegiline (1 mg/kg) was intraperitoneally administered once a day for 7 days after the surgery. PND, including impulsive-like behaviors and cognitive impairment, was evaluated by open field test, elevated plus maze, and fear conditioning. Thereafter, pathological changes of neurodegeneration were explored by western blot and immunofluorescence assays. RESULTS Selegiline administration significantly ameliorated TF-induced impulsive-like behaviors and reduced excessive GABA production in reactive hippocampal astrocytes. Moreover, astrocyte-specific NLRP3 knockout mice reversed TF-induced impulsive-like and cognitive impairment behaviors, decreased GABA levels in reactive astrocytes, ameliorated NLRP3-associated inflammatory responses during the early stage, and restored neuronal degeneration in the hippocampus. CONCLUSIONS Our findings suggest that anesthesia and surgical procedures trigger neuroinflammation and cognitive deficits, which may be due to NLRP3-GABA activation in the hippocampus of aged mice.
Collapse
Affiliation(s)
- Lu-Ying Wang
- Department of Anesthesia and Trauma Research, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Xu-Peng Wang
- Department of Anesthesiology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jin-Meng Lv
- Department of Anesthesia and Trauma Research, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Yu-Dong Shan
- Department of Anesthesia and Trauma Research, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Shi-Yan Jia
- Department of Anesthesia and Trauma Research, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Zhi-Fang Yu
- Department of Anesthesia and Trauma Research, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Hui-Tao Miao
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Yue Xin
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
| |
Collapse
|
18
|
Dong F, Yan W, Meng Q, Song X, Cheng B, Liu Y, Yao R. Ebselen alleviates white matter lesions and improves cognitive deficits by attenuating oxidative stress via Keap1/Nrf2 pathway in chronic cerebral hypoperfusion mice. Behav Brain Res 2023; 448:114444. [PMID: 37098387 DOI: 10.1016/j.bbr.2023.114444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/02/2023] [Accepted: 03/20/2023] [Indexed: 04/27/2023]
Abstract
Oxidative stress is crucial in cerebral white matter lesions (WMLs) induced by chronic cerebral hypoperfusion. Therefore, ameliorating oxidative damage is considered to be a beneficial strategy for the treatment of WMLs. Ebselen (EbSe), a small lipid organoselenium compound, its lipid peroxidation activity is mediated through the glutathione peroxidase-mimetic properties. This study aimed to investigate the role of EbSe in WMLs after bilateral common carotid artery stenosis (BCAS). The BCAS model can moderately reduce cerebral blood flow, and mimics white matter damage caused by chronic cerebral hypoperfusion or small vessel disease. Laser Speckle Contrast Imaging (LSCI) was used to monitor the cerebral blood flow of mice. The spatial learning and memory were tested by using the eight-arm maze. LFB staining was used to detect demyelination. The expression of MBP, GFAP and Iba1 was assayed by immunofluorescence. The demyelination was assessed by Transmission Electron Microscope (TEM). The activities of MDA, SOD and GSH-Px were detected by assay kits. The mRNA levels of SOD, GSH-Px and HO-1 was detected by realtime PCR. The activation of the Nrf2/ARE pathway and the expression of SOD, GSH-Px and HO-1was assessed by Western blot. EbSe ameliorated cognitive deficits and white matter lesions induced by bilateral common carotid artery stenosis (BCAS). The expression of GFAP and Iba1 was decreased in the corpus callosum of BCAS mice after EbSe treatment. Moreover, EbSe alleviated the level of MDA by elevating the expression and mRNA of SOD, GSH-Px and HO-1 in BCAS mice. Furthermore, EbSe promoted the dissociation of the Keap1/Nrf2 complex, resulting in the accumulation of Nrf2 in the nucleus. This study demonstrates a favorable effect of EbSe on cognitive impairment in a chronic cerebral hypoperfusion model, and the improvement of EbSe's antioxidant property is mediated by Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Fuxing Dong
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China; Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Weixing Yan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Qiqi Meng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Xueli Song
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Bing Cheng
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Yaping Liu
- Laboratory of National Experimental Teaching and Demonstration Center of Basic Medicine, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu Province, China.
| |
Collapse
|
19
|
Davari S, D'Costa N, Ramezan R, Mielke JG. Chronic Early-Life Social Isolation Enhances Spatial Memory in Male and Female Rats. Behav Brain Res 2023; 447:114433. [PMID: 37037406 DOI: 10.1016/j.bbr.2023.114433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
Social adversity during childhood and adolescence can alter brain development in ways that may increase the likelihood of many prominent mental illnesses. To determine the underlying mechanisms, several animal models have been developed, such as Chronic Early-Life Social Isolation (CELSI), which sees rats isolated for several weeks after weaning. Although such a paradigm does cause many consistent changes in adult behaviour, one area where uncertainty exists concerns its effect upon hippocampal-dependent learning and memory. To help sort out how CELSI affects spatial learning and memory, male and female siblings from 15 Sprague-Dawley rat litters were stratified by sex and then randomly assigned to either group-housing (3 animals/cage), or social isolation (1 animal/cage) for 7 weeks. Spatial learning and memory were then tested over 5 days using the Morris water maze. Next, the animals were euthanised, and stress-sensitive biometrics, including serum corticosterone levels, were collected. Lastly, to determine whether CELSI affected neural cell density, the expression of key neuronal and glial proteins (such as PSD-95 and GFAP, respectively) was assessed in isolated hippocampal tissue using immunoblotting. Notably, both male and female rats that had experienced post-weaning social isolation displayed stronger spatial learning and memory abilities than their group-housed counterparts. As well, socially isolated male rats exhibited a clear increase in expression of PSD-95. However, housing condition did not seem to affect either stress-sensitive biometrics, or hippocampal GFAP expression. Our results support the possibility that CELSI may enhance some aspects of hippocampal-dependent behaviour in a fashion similar among male and female rats.
Collapse
Affiliation(s)
- Saeideh Davari
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Nicole D'Costa
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada
| | - Reza Ramezan
- Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, ON, Canada
| | - John G Mielke
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
20
|
Lee H, Ciabatti E, González-Rueda A, Williams E, Nugent F, Mookerjee S, Morgese F, Tripodi M. Combining long-term circuit mapping and network transcriptomics with SiR-N2c. Nat Methods 2023; 20:580-589. [PMID: 36864202 PMCID: PMC7614628 DOI: 10.1038/s41592-023-01787-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 01/23/2023] [Indexed: 03/04/2023]
Abstract
An exciting frontier in circuit neuroscience lies at the intersection between neural network mapping and single-cell genomics. Monosynaptic rabies viruses provide a promising platform for the merger of circuit mapping methods with -omics approaches. However, three key limitations have hindered the extraction of physiologically meaningful gene expression profiles from rabies-mapped circuits: inherent viral cytotoxicity, high viral immunogenicity and virus-induced alteration of cellular transcriptional regulation. These factors alter the transcriptional and translational profiles of infected neurons and their neighboring cells. To overcome these limitations we applied a self-inactivating genomic modification to the less immunogenic rabies strain, CVS-N2c, to generate a self-inactivating CVS-N2c rabies virus (SiR-N2c). SiR-N2c not only eliminates undesired cytotoxic effects but also substantially reduces gene expression alterations in infected neurons and dampens the recruitment of innate and acquired immune responses, thus enabling open-ended interventions on neural networks and their genetic characterization using single-cell genomic approaches.
Collapse
Affiliation(s)
- Hassal Lee
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ernesto Ciabatti
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| | | | - Elena Williams
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Fiona Nugent
- IMAXT Laboratory, Cancer Research UK Cambridge Institute, Cambridge, UK
| | | | - Fabio Morgese
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Marco Tripodi
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
21
|
Young JW, Kenton JA, Milienne-Petiot M, Deben D, Achim C, Geyer MA, Perry W, Grant IE, Minassian A. Chronic methamphetamine exposure exerts few effects on the iTat mouse model of HIV, but blocks Tat expression-induced slowed reward retrieval. Behav Brain Res 2023; 437:114109. [PMID: 36108778 PMCID: PMC10878174 DOI: 10.1016/j.bbr.2022.114109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 10/14/2022]
Abstract
Human immunodeficiency virus (HIV) continues to infect millions worldwide, negatively impacting neurobehavioral function. Further understanding of the combined effects of HIV and methamphetamine use is crucial, as methamphetamine use is prevalent in people with HIV. The HIV-associated protein Tat may contribute to cognitive dysfunction, modeled preclinically in mice using doxycycline (DOX)-inducible Tat expression (iTat). Tat may exert its effects on cognitive function via disruption of the dopamine transporter, similar to the action of methamphetamine. Additionally, Tat and methamphetamine both decrease interneuron populations, including those expressing calbindin. It is important to understand the combined effects of Tat and methamphetamine in preclinical models of HIV infection. Here, we used iTat transgenic mice and a chronic binge regimen of methamphetamine exposure to determine their combined impact on reward learning and motivation. We also measured calbindin expression in behavior-relevant brain regions. Before induction with DOX, iTat mice exhibited no differences in behavior. Chronic methamphetamine exposure before Tat induction impaired initial reward learning but did not affect motivation. Furthermore, DOX-induced Tat expression did not alter behavior, but slowed latencies to retrieve rewards. This effect of Tat, however, was not observed in methamphetamine-treated mice, indicative of a potential protective effect. Finally, Tat expression was associated with an increase in calbindin-expressing cells in the VTA, while methamphetamine exposure did not alter calbindin numbers. These findings may indicate a protective role of methamphetamine in HIV neuropathology, which in turn may help in our understanding of why people with HIV use methamphetamine at disproportionately higher rates.
Collapse
Affiliation(s)
- Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States.
| | - Johnny A Kenton
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | | | - Debbie Deben
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Cristian Achim
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States
| | - William Perry
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Igor E Grant
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; Research Service, VA San Diego Healthcare System, San Diego, CA, United States; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; VA Center of Excellence for Stress and Mental Health, Veterans Administration San Diego HealthCare System, 3350 La Jolla Village Drive, San Diego, CA, United States
| | - Arpi Minassian
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States; VA Center of Excellence for Stress and Mental Health, Veterans Administration San Diego HealthCare System, 3350 La Jolla Village Drive, San Diego, CA, United States
| |
Collapse
|
22
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
23
|
Zamani N, Osgoei LT, Aliaghaei A, Zamani N, Hassanian-Moghaddam H. Chronic exposure to methadone induces activated microglia and astrocyte and cell death in the cerebellum of adult male rats. Metab Brain Dis 2023; 38:323-338. [PMID: 36287354 DOI: 10.1007/s11011-022-01108-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 02/03/2023]
Abstract
Methadone is a centrally-acting synthetic opioid analgesic widely used in the methadone maintenance therapy (MMT) programs throughout the world. Considering its neurotoxic effects particularly on the cerebellum, this study aims to address the behavioral and histological alterations in the cerebellar cortex associated with methadone administration. Twenty-four adult male albino rats were randomized into two groups of control and methadone treatment. Methadone was subcutaneously administered (2.5-10 mg/kg) once a day for two consecutive weeks. The functional and structural changes in the cerebellum were compared to the control group. Our data revealed that treating rats with methadone not only induced cerebellar atrophy, but also prompted the actuation of microgliosis, astrogliosis, and apoptotic biomarkers. We further demonstrated that treating rats with methadone increased complexity of astrocyte processes and decreased complexity of microglia processes. Our result showed that methadone impaired motor coordination and locomotor performance and neuromuscular activity. Additionally, relative gene expression of TNF-α, caspase-3 and RIPK3 increased significantly due to methadone. Our findings suggest that methadone administration has a neurodegenerative effect on the cerebellar cortex via dysregulation of microgliosis, astrogliosis, apoptosis, and neuro-inflammation.
Collapse
Affiliation(s)
- Naghmeh Zamani
- Department of Biology, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Laya Takbiri Osgoei
- Department of Microbiology, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nasim Zamani
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hassanian-Moghaddam
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
She X, Zhou C, Liang Z, Xie J, Zhao S, Tao J, Zhang Y, Mao J, Chen Y, Shen L. Hypodense regions in the peripapillary region increased the risk of macular retinoschisis detected by optical coherence tomography. Front Med (Lausanne) 2022; 9:1018580. [DOI: 10.3389/fmed.2022.1018580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
PurposeThe purpose of the present study was to investigate the clinical features of peripapillary regions in patients with myopic macular retinoschisis (MRS) and its association with the development of retinoschisis (RS).MethodsIn this cross-sectional study, high-myopic patients with or without MRS were recruited, and the hypodense regions were analyzed in the peripapillary regions. The vitreoretinal adhesions around both macular and paravascular arcades were compared between groups. The risk factors for the development of MRS were analyzed by logistic regression.ResultsOf 88 myopic eyes, MRS was detected in 45 eyes (51%). The eyes with MRS showed a higher rate of peripapillary and paravascular retinoschisis (P < 0.001 and P = 0.006). Hypodense regions were detected in 25 eyes (20.35%). Higher rates of horizontal and vertical macular MRS were detected in the hypodense group (P = 0.012 and P = 0.002). Lower refractive error, longer axial length, and higher rates of outer retinoschisis both in horizontal and vertical macular regions were observed in the hypodense group (P = 0.012, P = 0.006, P = 0.038, and P = 0.034). Higher rates of inner and outer retinoschisis, vitreoschisis, and microfolds along superior vascular arcade were detected in the hypodense group (P = 0.005, P = 0.001, P = 0.014, and P = 0.014). Higher rates of internal limiting membrane (ILM) detachment, inner and outer RS were detected along the inferior vascular arcade in the hypodense group (P = 0.008, P = 0.001, and P = 0.028). Hypodense regions, the axial length and PICC (peripapillary intrachoroidal cavitation) were significantly correlated with the severity of MRS (Odds ratio = 0.207, P = 0.010; Odds ratio = 1.399, P = 0.016; Odds ratio = 0.142, P = 0.010).ConclusionsThe hypodense regions were likely to affect outer retinoschisis both in macular and paravascular regions. It was a risk factor for the development of MRS.
Collapse
|
25
|
Decoding molecular programs in melanoma brain metastases. Nat Commun 2022; 13:7304. [PMID: 36435874 PMCID: PMC9701224 DOI: 10.1038/s41467-022-34899-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 11/07/2022] [Indexed: 11/28/2022] Open
Abstract
Melanoma brain metastases (MBM) variably respond to therapeutic interventions; thus determining patient's prognosis. However, the mechanisms that govern therapy response are poorly understood. Here, we use a multi-OMICS approach and targeted sequencing (TargetSeq) to unravel the programs that potentially control the development of progressive intracranial disease. Molecularly, the expression of E-cadherin (Ecad) or NGFR, the BRAF mutation state and level of immune cell infiltration subdivides tumors into proliferative/pigmented and invasive/stem-like/therapy-resistant irrespective of the intracranial location. The analysis of MAPK inhibitor-naive and refractory MBM reveals switching from Ecad-associated into NGFR-associated programs during progression. NGFR-associated programs control cell migration and proliferation via downstream transcription factors such as SOX4. Moreover, global methylome profiling uncovers 46 differentially methylated regions that discriminate BRAFmut and wildtype MBM. In summary, we propose that the expression of Ecad and NGFR sub- classifies MBM and suggest that the Ecad-to-NGFR phenotype switch is a rate-limiting process which potentially indicates drug-response and intracranial progression states in melanoma patients.
Collapse
|
26
|
Toledano-Díaz A, Álvarez MI, Toledano A. The relationships between neuroglial and neuronal changes in Alzheimer's disease, and the related controversies II: gliotherapies and multimodal therapy. J Cent Nerv Syst Dis 2022; 14:11795735221123896. [PMID: 36407561 PMCID: PMC9666878 DOI: 10.1177/11795735221123896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/05/2022] [Indexed: 08/30/2023] Open
Abstract
Since the original description of Alzheimer´s disease (AD), research into this condition has mainly focused on assessing the alterations to neurons associated with dementia, and those to the circuits in which they are involved. In most of the studies on human brains and in many models of AD, the glial cells accompanying these neurons undergo concomitant alterations that aggravate the course of neurodegeneration. As a result, these changes to neuroglial cells are now included in all the "pathogenic cascades" described in AD. Accordingly, astrogliosis and microgliosis, the main components of neuroinflammation, have been integrated into all the pathogenic theories of this disease, as discussed in this part of the two-part monograph that follows an accompanying article on gliopathogenesis and glioprotection. This initial reflection verified the implication of alterations to the neuroglia in AD, suggesting that these cells may also represent therapeutic targets to prevent neurodegeneration. In this second part of the monograph, we will analyze the possibilities of acting on glial cells to prevent or treat the neurodegeneration that is the hallmark of AD and other pathologies. Evidence of the potential of different pharmacological, non-pharmacological, cell and gene therapies (widely treated) to prevent or treat this disease is now forthcoming, in most cases as adjuncts to other therapies. A comprehensive AD multimodal therapy is proposed in which neuronal and neuroglial pharmacological treatments are jointly considered, as well as the use of new cell and gene therapies and non-pharmacological therapies that tend to slow down the progress of dementia.
Collapse
|
27
|
McCamy KM, Rees KA, Winzer-Serhan UH. Peripheral immune challenges elicit differential up-regulation of hippocampal cytokine and chemokine mRNA expression in a mouse model of the 15q13.3 microdeletion syndrome. Cytokine 2022; 159:156005. [PMID: 36084604 DOI: 10.1016/j.cyto.2022.156005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/03/2022]
Abstract
The human heterozygous 15q13.3 microdeletion is associated with neuropathological disorders, most prominently with epilepsy and intellectual disability. The 1.5 Mb deletion encompasses six genes (FAN1 [MTMR15], MTMR10, TRPM1, KLF13, OTUD7A, and CHRNA7); all but one (TRPM1) are expressed in the brain. The 15q13.3 microdeletion causes highly variable neurological symptoms, and confounding factors may contribute to a more severe phenotype. CHRNA7 and KLF13 are involved in immune system regulation and altered immune responses may contribute to neurological deficits. We used the Df[h15q13]/+ transgenic mouse model with a heterozygous deletion of the orthologous region (Het) to test the hypothesis that the microdeletion increases innate immune responses compared to wild type (WT). Male and female mice were acutely challenged with the bacteriomimetic lipopolysaccharide (LPS, 0.1 mg/kg, i.p.) or the viral mimetic polyinosinic:polycytidylic acid (Poly(I:C), 5 mg/kg). Hippocampal mRNA expression of pro-inflammatory cytokines and chemokines were determined three hours after injection using quantitative PCR analysis. In controls, expression was not affected by sex or genotype. LPS and Poly(I:C) resulted in significantly increased hippocampal expression of cytokines, chemokines, and interferon-γ (IFNγ), with more robust increases for TNF-α, IL-6, IL-1β, CXCL1, and CCL2 by LPS, higher induction of IFNγ by Poly(I:C), and similar increases of CCL4 and CCL5 by both agents. Generally, Hets exhibited stronger responses than WT mice, and significant effects of genotype or genotype × treatment interactions were detected for CXCL1 and CCL5, and IL-6, IL-1β, and CCL4, respectively, after LPS. Sex differences were detected for some targets. LPS but not Poly(I:C), reduced overnight burrowing independent of sex or genotype, suggesting that LPS induced sickness behavior. Thus, mice carrying the microdeletion have an increased innate immune response following a LPS challenge, but further studies will have to determine the extent and mechanisms of altered immune activation and subsequent contributions to 15q13.3 microdeletion associated deficits.
Collapse
Affiliation(s)
- Kristin M McCamy
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Katherine A Rees
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Ursula H Winzer-Serhan
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States.
| |
Collapse
|
28
|
Zhang X, Wolfinger A, Wu X, Alnafisah R, Imami A, Hamoud AR, Lundh A, Parpura V, McCullumsmith RE, Shukla R, O’Donovan SM. Gene Enrichment Analysis of Astrocyte Subtypes in Psychiatric Disorders and Psychotropic Medication Datasets. Cells 2022; 11:3315. [PMID: 36291180 PMCID: PMC9600295 DOI: 10.3390/cells11203315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/26/2022] Open
Abstract
Astrocytes have many important functions in the brain, but their roles in psychiatric disorders and their responses to psychotropic medications are still being elucidated. Here, we used gene enrichment analysis to assess the relationships between different astrocyte subtypes, psychiatric diseases, and psychotropic medications (antipsychotics, antidepressants and mood stabilizers). We also carried out qPCR analyses and "look-up" studies to assess the chronic effects of these drugs on astrocyte marker gene expression. Our bioinformatic analysis identified gene enrichment of different astrocyte subtypes in psychiatric disorders. The highest level of enrichment was found in schizophrenia, supporting a role for astrocytes in this disorder. We also found differential enrichment of astrocyte subtypes associated with specific biological processes, highlighting the complex responses of astrocytes under pathological conditions. Enrichment of protein phosphorylation in astrocytes and disease was confirmed by biochemical analysis. Analysis of LINCS chemical perturbagen gene signatures also found that kinase inhibitors were highly discordant with astrocyte-SCZ associated gene signatures. However, we found that common gene enrichment of different psychotropic medications and astrocyte subtypes was limited. These results were confirmed by "look-up" studies and qPCR analysis, which also reported little effect of psychotropic medications on common astrocyte marker gene expression, suggesting that astrocytes are not a primary target of these medications. Conversely, antipsychotic medication does affect astrocyte gene marker expression in postmortem schizophrenia brain tissue, supporting specific astrocyte responses in different pathological conditions. Overall, this study provides a unique view of astrocyte subtypes and the effect of medications on astrocytes in disease, which will contribute to our understanding of their role in psychiatric disorders and offers insights into targeting astrocytes therapeutically.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Alyssa Wolfinger
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Xiaojun Wu
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Rawan Alnafisah
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Ali Imami
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Abdul-rizaq Hamoud
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Anna Lundh
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert E. McCullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
- Promedica Neurosciences Institute, Toledo, OH 43606, USA
| | - Rammohan Shukla
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
| | | |
Collapse
|
29
|
Bastelica P, Labbé A, El Maftouhi A, Hamard P, Paques M, Baudouin C. Rôle de la lame criblée dans la pathogenèse du glaucome. Une revue de la littérature. J Fr Ophtalmol 2022; 45:952-966. [DOI: 10.1016/j.jfo.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
|
30
|
Astroglial and oligodendroglial markers in the cuprizone animal model for de- and remyelination. Histochem Cell Biol 2022; 158:15-38. [PMID: 35380252 PMCID: PMC9246805 DOI: 10.1007/s00418-022-02096-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2022] [Indexed: 01/08/2023]
Abstract
Myelin loss with consecutive axon degeneration and impaired remyelination are the underlying causes of progressive disease in patients with multiple sclerosis. Astrocytes are suggested to play a major role in these processes. The unmasking of distinct astrocyte identities in health and disease would help to understand the pathophysiological mechanisms in which astrocytes are involved. However, the number of specific astrocyte markers is limited. Therefore, we performed immunohistochemical studies and analyzed various markers including GFAP, vimentin, S100B, ALDH1L1, and LCN2 during de- and remyelination using the toxic murine cuprizone animal model. Applying this animal model, we were able to confirm overlapping expression of vimentin and GFAP and highlighted the potential of ALDH1L1 as a pan-astrocytic marker, in agreement with previous data. Only a small population of GFAP-positive astrocytes in the corpus callosum highly up-regulated LCN2 at the peak of demyelination and S100B expression was found in a subset of oligodendroglia as well, thus S100B turned out to have a limited use as a particular astroglial marker. Additionally, numerous GFAP-positive astrocytes in the lateral corpus callosum did not express S100B, further strengthening findings of heterogeneity in the astrocytic population. In conclusion, our results acknowledged that GFAP, vimentin, LCN2, and ALDH1L1 serve as reliable marker to identify activated astrocytes during cuprizone-induced de- and remyelination. Moreover, there were clear regional and temporal differences in protein and mRNA expression levels and patterns of the studied markers, generally between gray and white matter structures.
Collapse
|
31
|
Masrori P, Beckers J, Gossye H, Van Damme P. The role of inflammation in neurodegeneration: novel insights into the role of the immune system in C9orf72 HRE-mediated ALS/FTD. Mol Neurodegener 2022; 17:22. [PMID: 35303907 PMCID: PMC8932121 DOI: 10.1186/s13024-022-00525-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/25/2022] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation is an important hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). An inflammatory reaction to neuronal injury is deemed vital for neuronal health and homeostasis. However, a continued activation of the inflammatory response can be detrimental to remaining neurons and aggravate the disease process. Apart from a disease modifying role, some evidence suggests that neuroinflammation may also contribute to the upstream cause of the disease. In this review, we will first focus on the role of neuroinflammation in the pathogenesis of chromosome 9 open reading frame 72 gene (C9orf72) hexanucleotide repeat expansions (HRE)-mediated ALS/FTD (C9-ALS/FTD). Additionally, we will discuss evidence from ex vivo and in vivo studies and finally, we briefly summarize the trials and progress of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Pegah Masrori
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000, Leuven, Belgium.,Laboratory of Neurobiology, Experimental Neurology, Center for Brain and Disease Research, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, 602, 3000, Leuven, PB, Belgium.,Neurology Department, University Hospitals Leuven, Campus Gasthuisberg, Herestraat 49, 3000, Leuven, Belgium.,Department of Neurology, University Hospital Antwerp, 2650, Edegem, Belgium
| | - Jimmy Beckers
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000, Leuven, Belgium.,Laboratory of Neurobiology, Experimental Neurology, Center for Brain and Disease Research, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, 602, 3000, Leuven, PB, Belgium
| | - Helena Gossye
- Department of Neurology, University Hospital Antwerp, 2650, Edegem, Belgium.,VIB Center for Molecular Neurology, Neurodegenerative Brain Diseases, University of Antwerp, 2000, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, 2000, Antwerp, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000, Leuven, Belgium. .,Laboratory of Neurobiology, Experimental Neurology, Center for Brain and Disease Research, VIB, Campus Gasthuisberg, O&N5, Herestraat 49, 602, 3000, Leuven, PB, Belgium. .,Neurology Department, University Hospitals Leuven, Campus Gasthuisberg, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
32
|
Yu A, Wang S, Xing Y, Han M, Shao K. 7,8-Dihydroxyflavone alleviates apoptosis and inflammation induced by retinal ischemia-reperfusion injury via activating TrkB/Akt/NF-kB signaling pathway. Int J Med Sci 2022; 19:13-24. [PMID: 34975295 PMCID: PMC8692126 DOI: 10.7150/ijms.65733] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/26/2021] [Indexed: 12/31/2022] Open
Abstract
Retinal ischemia-reperfusion injury (RIRI) is of common occurrence in retinal and optic nerve diseases. The BDNF/TrkB signaling pathway has been examined to be neuroprotective in RIRI. In this study, we investigated the role of a potent selective TrkB agonist 7,8-dihydroxyfavone (DHF) in rat retinas with RIRI. Our results showed that RIRI inhibited the conversion of BDNF precursor (proBDNF) to mature BDNF (mBDNF) and increased the level of neuronal cell apoptosis. Compared with RIRI, DHF+RIRI reduced proBDNF level and at the same time increased mBDNF level. Moreover, DHF administration effectively activated TrkB signaling and and downstream Akt and Erk signaling pathways which increased nerve cell survival. The combined effects of mBDNF/proBDNF increase and TrkB signaling activation lead to reduction of apoptosis level and protection of retinas with RIRI. Moreover, it was also found that astrocytes labeled by GFAP were activated in RIRI and NF-kB mediated the increased expressions of inflammatory factors and these effects were partially reversed by DHF administration. Besides, we also used RNA sequencing to analyze the differently expressed genes (DEGs) and their enriched (Kyoto Encyclopedia of Genes and Genomes) KEGG pathways between Sham, RIRI, and DHF+RIRI. It was found that 1543 DEGs were differently expressed in RIRI and 619 DEGs were reversed in DHF+RIRI. The reversed DEGs were typically enriched in PI3K-Akt signaling pathway, Jak-STAT signaling pathway, NF-kB signaling pathway, and Apoptosis. To sum up, the DHF administration alleviated apoptosis and inflammation induced by RIRI via activating TrkB signaling pathway and may serve as a promising drug candidate for RIRI related ophthalmopathy.
Collapse
Affiliation(s)
- Aihua Yu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, Hubei Province, China
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan University,Wuhan 430071, Hubei Province, China
| | - Shun Wang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, Hubei Province, China
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan University,Wuhan 430071, Hubei Province, China
| | - Yiqiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, Hubei Province, China
| | - Mengyao Han
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan University,Wuhan 430071, Hubei Province, China
| | - Kun Shao
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan University,Wuhan 430071, Hubei Province, China
| |
Collapse
|
33
|
The neuroprotective function of 2-carba-cyclic phosphatidic acid: Implications for tenascin-C via astrocytes in traumatic brain injury. J Neuroimmunol 2021; 361:577749. [PMID: 34688067 DOI: 10.1016/j.jneuroim.2021.577749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023]
Abstract
We examined the mechanism how 2-carba-cyclic phosphatidic acid (2ccPA), a lipid mediator, regulates neuronal apoptosis in traumatic brain injury (TBI). First, we found 2ccPA suppressed neuronal apoptosis after the injury, and increased the immunoreactivity of tenascin-C (TN-C), an extracellular matrix protein by 2ccPA in the vicinity of the wound region. 2ccPA increased the mRNA expression levels of Tnc in primary cultured astrocytes, and the conditioned medium of 2ccPA-treated astrocytes suppressed the apoptosis of cortical neurons. The neuroprotective effect of TN-C was abolished by knockdown of TN-C. These results indicate that 2ccPA contributes to neuroprotection via TN-C from astrocytes in TBI.
Collapse
|
34
|
Oligodendrocytes depend on MCL-1 to prevent spontaneous apoptosis and white matter degeneration. Cell Death Dis 2021; 12:1133. [PMID: 34873168 PMCID: PMC8648801 DOI: 10.1038/s41419-021-04422-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 11/26/2022]
Abstract
Neurologic disorders often disproportionately affect specific brain regions, and different apoptotic mechanisms may contribute to white matter pathology in leukodystrophies or gray matter pathology in poliodystrophies. We previously showed that neural progenitors that generate cerebellar gray matter depend on the anti-apoptotic protein BCL-xL. Conditional deletion of Bcl-xL in these progenitors produces spontaneous apoptosis and cerebellar hypoplasia, while similar conditional deletion of Mcl-1 produces no phenotype. Here we show that, in contrast, postnatal oligodendrocytes depend on MCL-1. We found that brain-wide Mcl-1 deletion caused apoptosis specifically in mature oligodendrocytes while sparing astrocytes and oligodendrocyte precursors, resulting in impaired myelination and progressive white matter degeneration. Disabling apoptosis through co-deletion of Bax or Bak rescued white matter degeneration, implicating the intrinsic apoptotic pathway in Mcl-1-dependence. Bax and Bak co-deletions rescued different aspects of the Mcl-1-deleted phenotype, demonstrating their discrete roles in white matter stability. MCL-1 protein abundance was reduced in eif2b5-mutant mouse model of the leukodystrophy vanishing white matter disease (VWMD), suggesting the potential for MCL-1 deficiency to contribute to clinical neurologic disease. Our data show that oligodendrocytes require MCL-1 to suppress apoptosis, implicate MCL-1 deficiency in white matter pathology, and suggest apoptosis inhibition as a leukodystrophy therapy.
Collapse
|
35
|
Antimony-induced astrocyte activation via mitogen-activated protein kinase activation-dependent CREB phosphorylation. Toxicol Lett 2021; 352:9-16. [PMID: 34571074 DOI: 10.1016/j.toxlet.2021.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/06/2021] [Accepted: 09/21/2021] [Indexed: 11/23/2022]
Abstract
Recent studies suggest that the chemical element antimony (Sb) is neurotoxic; however, the molecular mechanisms behind Sb-related neuronal damage are currently unknown. In this study, we found that Sb exposure promoted astrocyte proliferation and increased the expression of inducible nitric oxide synthase (iNOS) and glial fibrillary acidic protein (GFAP), two key protein markers of reactive astrogliosis, at both the gene and protein level, suggesting that Sb induced astrocyte activation. Moreover, the p38 mitogen-activated protein kinase (p38 MAPK) and extracellular signal-related kinase (ERK) pathways were activated following Sb exposure. Inhibition of p38 MAPK reduced Sb-induced iNOS and GFAP upregulation, while inhibiting ERK reduced GFAP expression only, in Sb-exposed C6 cells. Sb treatment also induced the phosphorylation of cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB), and the inhibition of CREB caused a reduction in Sb-induced GFAP and iNOS expression. Furthermore, inhibiting both p38 MAPK and ERK effectively alleviated CREB phosphorylation in Sb-exposed C6 cells. Taken together, our results suggest that p38 MAPK and ERK activation mediate Sb-induced astrocyte activation through CREB phosphorylation. These results help to clarify the molecular mechanisms underlying Sb-associated neurotoxicity.
Collapse
|
36
|
Mirzaei N, Davis N, Chau TW, Sastre M. Astrocyte Reactivity in Alzheimer's Disease: Therapeutic Opportunities to Promote Repair. Curr Alzheimer Res 2021; 19:1-15. [PMID: 34719372 DOI: 10.2174/1567205018666211029164106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/02/2021] [Accepted: 07/31/2021] [Indexed: 11/22/2022]
Abstract
Astrocytes are fast climbing the ladder of importance in neurodegenerative disorders, particularly in Alzheimer's disease (AD), with the prominent presence of reactive astrocytes sur- rounding amyloid β- plaques, together with activated microglia. Reactive astrogliosis, implying morphological and molecular transformations in astrocytes, seems to precede neurodegeneration, suggesting a role in the development of the disease. Single-cell transcriptomics has recently demon- strated that astrocytes from AD brains are different from "normal" healthy astrocytes, showing dys- regulations in areas such as neurotransmitter recycling, including glutamate and GABA, and im- paired homeostatic functions. However, recent data suggest that the ablation of astrocytes in mouse models of amyloidosis results in an increase in amyloid pathology as well as in the inflammatory profile and reduced synaptic density, indicating that astrocytes mediate neuroprotective effects. The idea that interventions targeting astrocytes may have great potential for AD has therefore emerged, supported by a range of drugs and stem cell transplantation studies that have successfully shown a therapeutic effect in mouse models of AD. In this article, we review the latest reports on the role and profile of astrocytes in AD brains and how manipulation of astrocytes in animal mod- els has paved the way for the use of treatments enhancing astrocytic function as future therapeutic avenues for AD.
Collapse
Affiliation(s)
- Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048. United States
| | - Nicola Davis
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| | - Tsz Wing Chau
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| |
Collapse
|
37
|
Moulson AJ, Squair JW, Franklin RJM, Tetzlaff W, Assinck P. Diversity of Reactive Astrogliosis in CNS Pathology: Heterogeneity or Plasticity? Front Cell Neurosci 2021; 15:703810. [PMID: 34381334 PMCID: PMC8349991 DOI: 10.3389/fncel.2021.703810] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
Astrocytes are essential for the development and homeostatic maintenance of the central nervous system (CNS). They are also critical players in the CNS injury response during which they undergo a process referred to as "reactive astrogliosis." Diversity in astrocyte morphology and gene expression, as revealed by transcriptional analysis, is well-recognized and has been reported in several CNS pathologies, including ischemic stroke, CNS demyelination, and traumatic injury. This diversity appears unique to the specific pathology, with significant variance across temporal, topographical, age, and sex-specific variables. Despite this, there is limited functional data corroborating this diversity. Furthermore, as reactive astrocytes display significant environmental-dependent plasticity and fate-mapping data on astrocyte subsets in the adult CNS is limited, it remains unclear whether this diversity represents heterogeneity or plasticity. As astrocytes are important for neuronal survival and CNS function post-injury, establishing to what extent this diversity reflects distinct established heterogeneous astrocyte subpopulations vs. environmentally dependent plasticity within established astrocyte subsets will be critical for guiding therapeutic development. To that end, we review the current state of knowledge on astrocyte diversity in the context of three representative CNS pathologies: ischemic stroke, demyelination, and traumatic injury, with the goal of identifying key limitations in our current knowledge and suggesting future areas of research needed to address them. We suggest that the majority of identified astrocyte diversity in CNS pathologies to date represents plasticity in response to dynamically changing post-injury environments as opposed to heterogeneity, an important consideration for the understanding of disease pathogenesis and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Aaron J. Moulson
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| | - Jordan W. Squair
- Department of Clinical Neuroscience, Faculty of Life Sciences, Center for Neuroprosthetics and Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), NeuroRestore, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Robin J. M. Franklin
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Peggy Assinck
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Neudecker V, Perez-Zoghbi JF, Martin LD, Dissen GA, Grafe MR, Brambrink AM. Astrogliosis in juvenile non-human primates 2 years after infant anaesthesia exposure. Br J Anaesth 2021; 127:447-457. [PMID: 34266661 DOI: 10.1016/j.bja.2021.04.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Infant anaesthesia causes acute brain cell apoptosis, and later in life cognitive deficits and behavioural alterations, in non-human primates (NHPs). Various brain injuries and neurodegenerative conditions are characterised by chronic astrocyte activation (astrogliosis). Glial fibrillary acidic protein (GFAP), an astrocyte-specific protein, increases during astrogliosis and remains elevated after an injury. Whether infant anaesthesia is associated with a sustained increase in GFAP is unknown. We hypothesised that GFAP is increased in specific brain areas of NHPs 2 yr after infant anaesthesia, consistent with prior injury. METHODS Eight 6-day-old NHPs per group were exposed to 5 h isoflurane once (1×) or three times (3×), or to room air as a control (Ctr). Two years after exposure, their brains were assessed for GFAP density changes in the primary visual cortex (V1), perirhinal cortex (PRC), hippocampal subiculum, amygdala, and orbitofrontal cortex (OFC). We also assessed concomitant microglia activation and hippocampal neurogenesis. RESULTS Compared with controls, GFAP densities in V1 were increased in exposed groups (Ctr: 0.208 [0.085-0.427], 1×: 0.313 [0.108-0.533], 3×: 0.389 [0.262-0.652]), whereas the density of activated microglia was unchanged. In addition, GFAP densities were increased in the 3× group in the PRC and the subiculum, and in both exposure groups in the amygdala, but there was no increase in the OFC. There were no differences in hippocampal neurogenesis among groups. CONCLUSIONS Two years after infant anaesthesia, NHPs show increased GFAP without concomitant microglia activation in specific brain areas. These long-lasting structural changes in the brain caused by infant anaesthesia exposure may be associated with functional alterations at this age.
Collapse
Affiliation(s)
- Viola Neudecker
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Jose F Perez-Zoghbi
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Lauren D Martin
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Gregory A Dissen
- Division of Comparative Medicine, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Marjorie R Grafe
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Ansgar M Brambrink
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
39
|
Nutraceuticals and Enteric Glial Cells. Molecules 2021; 26:molecules26123762. [PMID: 34205534 PMCID: PMC8234579 DOI: 10.3390/molecules26123762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
Until recently, glia were considered to be a structural support for neurons, however further investigations showed that glial cells are equally as important as neurons. Among many different types of glia, enteric glial cells (EGCs) found in the gastrointestinal tract, have been significantly underestimated, but proved to play an essential role in neuroprotection, immune system modulation and many other functions. They are also said to be remarkably altered in different physiopathological conditions. A nutraceutical is defined as any food substance or part of a food that provides medical or health benefits, including prevention and treatment of the disease. Following the description of these interesting peripheral glial cells and highlighting their role in physiological and pathological changes, this article reviews all the studies on the effects of nutraceuticals as modulators of their functions. Currently there are only a few studies available concerning the effects of nutraceuticals on EGCs. Most of them evaluated molecules with antioxidant properties in systemic conditions, whereas only a few studies have been performed using models of gastrointestinal disorders. Despite the scarcity of studies on the topic, all agree that nutraceuticals have the potential to be an interesting alternative in the prevention and/or treatment of enteric gliopathies (of systemic or local etiology) and their associated gastrointestinal conditions.
Collapse
|
40
|
Neuroprotection in Glaucoma: NAD +/NADH Redox State as a Potential Biomarker and Therapeutic Target. Cells 2021; 10:cells10061402. [PMID: 34198948 PMCID: PMC8226607 DOI: 10.3390/cells10061402] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness worldwide. Its prevalence and incidence increase exponentially with age and the level of intraocular pressure (IOP). IOP reduction is currently the only therapeutic modality shown to slow glaucoma progression. However, patients still lose vision despite best treatment, suggesting that other factors confer susceptibility. Several studies indicate that mitochondrial function may underlie both susceptibility and resistance to developing glaucoma. Mitochondria meet high energy demand, in the form of ATP, that is required for the maintenance of optimum retinal ganglion cell (RGC) function. Reduced nicotinamide adenine dinucleotide (NAD+) levels have been closely correlated to mitochondrial dysfunction and have been implicated in several neurodegenerative diseases including glaucoma. NAD+ is at the centre of various metabolic reactions culminating in ATP production—essential for RGC function. In this review we present various pathways that influence the NAD+(H) redox state, affecting mitochondrial function and making RGCs susceptible to degeneration. Such disruptions of the NAD+(H) redox state are generalised and not solely induced in RGCs because of high IOP. This places the NAD+(H) redox state as a potential systemic biomarker for glaucoma susceptibility and progression; a hypothesis which may be tested in clinical trials and then translated to clinical practice.
Collapse
|
41
|
Chiareli RA, Carvalho GA, Marques BL, Mota LS, Oliveira-Lima OC, Gomes RM, Birbrair A, Gomez RS, Simão F, Klempin F, Leist M, Pinto MCX. The Role of Astrocytes in the Neurorepair Process. Front Cell Dev Biol 2021; 9:665795. [PMID: 34113618 PMCID: PMC8186445 DOI: 10.3389/fcell.2021.665795] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are highly specialized glial cells responsible for trophic and metabolic support of neurons. They are associated to ionic homeostasis, the regulation of cerebral blood flow and metabolism, the modulation of synaptic activity by capturing and recycle of neurotransmitters and maintenance of the blood-brain barrier. During injuries and infections, astrocytes act in cerebral defense through heterogeneous and progressive changes in their gene expression, morphology, proliferative capacity, and function, which is known as reactive astrocytes. Thus, reactive astrocytes release several signaling molecules that modulates and contributes to the defense against injuries and infection in the central nervous system. Therefore, deciphering the complex signaling pathways of reactive astrocytes after brain damage can contribute to the neuroinflammation control and reveal new molecular targets to stimulate neurorepair process. In this review, we present the current knowledge about the role of astrocytes in brain damage and repair, highlighting the cellular and molecular bases involved in synaptogenesis and neurogenesis. In addition, we present new approaches to modulate the astrocytic activity and potentiates the neurorepair process after brain damage.
Collapse
Affiliation(s)
| | | | | | - Lennia Soares Mota
- Department of Pharmacology, Federal University of Goias, Goiânia, Brazil
| | | | | | - Alexander Birbrair
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renato Santiago Gomez
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Simão
- Research Division, Vascular Cell Biology, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | | | - Marcel Leist
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
42
|
Chronic unpredictable stress negatively regulates hippocampal neurogenesis and promote anxious depression-like behavior via upregulating apoptosis and inflammatory signals in adult rats. Brain Res Bull 2021; 172:164-179. [PMID: 33895271 DOI: 10.1016/j.brainresbull.2021.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/28/2022]
Abstract
Psychological and physical stress play a pivotal role in etiology of anxiety and depression. Chronic psychological and physical stress modify various physiological phenomena, as a consequence of which oxidative stress, decreased neurotransmitter level, elevated corticosterone level and altered NSC homeostasis is observed. However, the precise mechanism by which chronic stress induce anxious depression and modify internal milieu is still unknown. Herein, we show that exposure to CUS increase oxidative stress, microgliosis, astrogliosis while it reduces hippocampal NSC proliferation, neuronal differentiation and maturation in adult rats. CUS exposure in rats reduce dopamine and serotonin level in cortex and hippocampus, which result in increased anxiety and depression-like phenotypes. We also found elevated level of NF-κB and TNF-α while decreased anti-inflammatory cytokine IL-10 level, that led to increased expression of Bax and cleaved Caspase-3 whereas down regulation of antiapoptotic protein Bcl2. Additionally, CUS altered adult hippocampal neurogenesis, increased gliosis and neuronal apoptosis in cerebral cortex and hippocampus which might be associated with reduced AKT and increased ERK signaling, as seen in the rat brain tissue. Taken together, these results indicate that CUS induce oxidative stress and neuroinflammation which directly affects NSC dynamics, monoamines levels and behavioral functions in adult rats.
Collapse
|
43
|
Navarro A, García M, Rodrigues AS, Garcia PV, Camarinho R, Segovia Y. Reactive astrogliosis in the dentate gyrus of mice exposed to active volcanic environments. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:213-226. [PMID: 33283687 DOI: 10.1080/15287394.2020.1850381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Air pollution has been associated with neuroinflammatory processes and is considered a risk factor for the development of neurodegenerative diseases. Volcanic environments are considered a natural source of air pollution. However, the effects of natural source air pollution on the central nervous system (CNS) have not been reported, despite the fact that up to 10% of the world's population lives near a historically active volcano. In order to assess the response of the CNS to such exposure, our study was conducted in the island of Sao Miguel (Azores, Portugal) in two different areas: Furnas, which is volcanically active one, and compared to Rabo de Peixe, a reference site without manifestations of active volcanism using Mus musculus as a bioindicator species. To evaluate the state of the astroglial population in the dentate gyrus in both samples, the number of astrocytes was determined using immunofluorescence methods (anti-GFAP and anti-GS). In addition, the astrocytic branches in that hippocampal area were examined. Our results showed an increase in GFAP+ astrocytes and a reduction in GS+ astrocytes in Furnas-exposed mice compared to animals from Rabo de Peixe. In addition, astrocytes in the dentate gyrus of chronically exposed animals exhibited longer branches compared to those residing at the reference site. Thus, reactive astrogliosis and astrocyte dysfunction are found in mice living in an active volcanic environment.
Collapse
Affiliation(s)
- A Navarro
- Department of Biotechnology, University of Alicante , Alicante, Spain
| | - M García
- Department of Biotechnology, University of Alicante , Alicante, Spain
| | - A S Rodrigues
- Faculty of Sciences and Technology, University of the Azores , Ponta Delgada, Portugal
- IVAR, Research Institute for Volcanology and Risk Assessment, University of the Azores , Ponta Delgada, Portugal
| | - P V Garcia
- Faculty of Sciences and Technology, University of the Azores , Ponta Delgada, Portugal
- cE3c, Centre for Ecology, Evolution and Environmental Changes, and Azorean Biodiversity Group, University of the Azores , Ponta Delgada, Portugal
| | - R Camarinho
- Faculty of Sciences and Technology, University of the Azores , Ponta Delgada, Portugal
- IVAR, Research Institute for Volcanology and Risk Assessment, University of the Azores , Ponta Delgada, Portugal
| | - Y Segovia
- Department of Biotechnology, University of Alicante , Alicante, Spain
| |
Collapse
|
44
|
Inhibition of ERK1/2 phosphorylation attenuates spinal cord injury induced astrocyte activation and inflammation through negatively regulating aquaporin-4 in rats. Brain Res Bull 2021; 170:162-173. [PMID: 33592275 DOI: 10.1016/j.brainresbull.2021.02.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 01/09/2023]
Abstract
The extracellular signal-regulated kinase (ERK) pathway has been reported to play a pivotal role in mediating spinal cord injury (SCI) progression. The present study aimed to investigate the effects of phosphorylated ERK1/2 (p-ERK1/2) inhibition on SCI-induced astrocyte activation and inflammation and its possible mechanism in rats. Here, female Sprague-Dawley rats were randomly assigned to four groups: (1) Sham group, (2) SCI group, (3) TGN-020 group (aquaporin-4, AQP4, blocking agent), (4) PD98059 group (ERK blocking agent). A well SCI model was established by compressing the thoracic vertebra 10 level (weight 35 g, time 5 min) in rats. Western blotting and immunofluorescence staining were used to measure the expression of associated proteins after SCI. HE staining and Nissl staining were performed to detect the morphological changes of spinal cords and the number of surviving neurons following SCI, respectively. The Basso-Beattie-Bresnahan open-field rating scale was used to evaluate functional locomotor recovery following SCI in rats. Our results demonstrated that SCI significantly induced the upregulation of aquaporin-4, p-ERK1/2, glial fibrillary acidic protein, proliferating cell nuclear antigen, and proinflammatory cytokines (tumor necrosis factor-α, interleukin-6 and interleukin-1β). However, treatment with TGN-020 or PD98059 could effectively inhibit astrocyte proliferation and proinflammatory cytokine release, preserve the number of surviving ventral horn neurons, and subsequently improve the locomotor function of rats after SCI. Interestingly, the SCI-induced elevation of AQP4 expression was downregulated by p-ERK1/2 inhibition, suggesting that blocking ERK1/2 phosphorylation could attenuate astrocyte activation and inflammatory processes through negative regulation of AQP4. Therefore, p-ERK1/2 blockade may be employed as a therapeutic target for SCI.
Collapse
|
45
|
Real CC, Suemoto CK, Binda KH, Grinberg LT, Pasqualucci CA, Jacob W, Ferretti-Rebustini REDL, Nitrini R, Leite REP, de Britto LR. Active lifestyle enhances protein expression profile in subjects with Lewy body pathology. Dement Neuropsychol 2021; 15:41-50. [PMID: 33907596 PMCID: PMC8049574 DOI: 10.1590/1980-57642021dn15-010004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Clinical trials of the effects of physical activity have reported improvements in symptoms and quality of life in patients with Parkinson's disease (PD). Additionally, morphological brain changes after exercising were reported in PD animal models. However, these lifestyle-related changes were not evaluated in postmortem brain tissue. OBJECTIVE We aimed to evaluate, by immunohistochemistry, astrocytes, tyrosine hydroxylase (TH) and structural proteins expression (neurofilaments and microtubules - MAP2) changes in postmortem brain samples of individuals with Lewy body pathology. METHODS Braak PD stage≥III samples, classified by neuropathology analysis, from The Biobank for Aging Studies were classified into active (n=12) and non-active (n=12) groups, according to physical activity lifestyle, and paired by age, sex and Braak staging. Substantia nigra and basal ganglia were evaluated. RESULTS Groups were not different in terms of age or gender and had similar PD neuropathological burden (p=1.00). We observed higher TH expression in the active group in the substantia nigra and the basal ganglia (p=0.04). Astrocytes was greater in the non-active subjects in the midbrain (p=0.03) and basal ganglia (p=0.0004). MAP2 levels were higher for non-active participants in the basal ganglia (p=0.003) and similar between groups in the substantia nigra (p=0.46). Neurofilament levels for non-active participants were higher in the substantia nigra (p=0.006) but not in the basal ganglia (p=0.24). CONCLUSION Active lifestyle seems to promote positive effects on brain by maintaining dopamine synthesis and structural protein expression in the nigrostriatal system and decrease astrogliosis in subjects with the same PD neuropathology burden.
Collapse
Affiliation(s)
- Caroline Cristiano Real
- Laboratoy of Nuclear Medicine, Faculdade de Medicina,
Universidade de São Paulo – São Paulo, SP, Brazil
- Laboratory of Cellular Neurobiology, Department of Physiology
and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo –
São Paulo, SP, Brazil
| | - Cláudia Kimie Suemoto
- Division of Geriatrics, Faculdade de Medicina, Universidade de
São Paulo – São Paulo, SP, Brazil
| | - Karina Henrique Binda
- Laboratory of Cellular Neurobiology, Department of Physiology
and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo –
São Paulo, SP, Brazil
| | - Lea Tenenholz Grinberg
- Memory and Aging Center, University of California San Francisco
– San Francisco, California, United States of America
- Department of Pathology, Faculdade de Medicina, Universidade de
São Paulo – São Paulo, SP, Brazil
| | | | - Wilson Jacob
- Division of Geriatrics, Faculdade de Medicina, Universidade de
São Paulo – São Paulo, SP, Brazil
| | | | - Ricardo Nitrini
- Department of Neurology, Faculdade de Medicina, Universidade de
São Paulo – São Paulo, SP, Brazil
| | | | - Luiz Roberto de Britto
- Laboratory of Cellular Neurobiology, Department of Physiology
and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo –
São Paulo, SP, Brazil
| |
Collapse
|
46
|
Du Preez A, Onorato D, Eiben I, Musaelyan K, Egeland M, Zunszain PA, Fernandes C, Thuret S, Pariante CM. Chronic stress followed by social isolation promotes depressive-like behaviour, alters microglial and astrocyte biology and reduces hippocampal neurogenesis in male mice. Brain Behav Immun 2021; 91:24-47. [PMID: 32755644 DOI: 10.1016/j.bbi.2020.07.015] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
Unpredictable chronic mild stress (UCMS) is one of the most commonly used, robust and translatable models for studying the neurobiological basis of major depression. Although the model currently has multiple advantages, it does not entirely follow the trajectory of the disorder, whereby depressive symptomology can often present months after exposure to stress. Furthermore, patients with depression are more likely to withdraw in response to their stressful experience, or as a symptom of their depression, and, in turn, this withdrawal/isolation can further exacerbate the stressful experience and the depressive symptomology. Therefore, we investigated the effect(s) of 6 weeks of UCMS followed by another 6 weeks of social isolation (referred to as UCMSI), on behaviour, corticosterone stress responsivity, immune system functioning, and hippocampal neurogenesis, in young adult male mice. We found that UCMSI induced several behavioural changes resembling depression but did not induce peripheral inflammation. However, UCMSI animals showed increased microglial activation in the ventral dentate gyrus (DG) of the hippocampus and astrocyte activation in both the dorsal and ventral DG, with increased GFAP-positive cell immunoreactivity, GFAP-positive cell hypertrophy and process extension, and increased s100β-positive cell density. Moreover, UCMSI animals had significantly reduced neurogenesis in the DG and reduced levels of peripheral vascular endothelial growth factor (VEGF) - a trophic factor produced by astrocytes and that stimulates neurogenesis. Finally, UCMSI mice also had normal baseline corticosterone levels but a smaller increase in corticosterone following acute stress, that is, the Porsolt Swim Test. Our work gives clinically relevant insights into the role that microglial and astrocyte functioning, and hippocampal neurogenesis may play in the context of stress, social isolation and depression, offering a potentially new avenue for therapeutic target.
Collapse
Affiliation(s)
- Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Diletta Onorato
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Inez Eiben
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Ksenia Musaelyan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Martin Egeland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
47
|
La Sala G, Di Pietro C, Matteoni R, Bolasco G, Marazziti D, Tocchini-Valentini GP. Gpr37l1/prosaposin receptor regulates Ptch1 trafficking, Shh production, and cell proliferation in cerebellar primary astrocytes. J Neurosci Res 2020; 99:1064-1083. [PMID: 33350496 DOI: 10.1002/jnr.24775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 02/24/2024]
Abstract
Mammalian cerebellar astrocytes critically regulate the differentiation and maturation of neuronal Purkinje cells and granule precursors. The G protein-coupled receptor 37-like 1 (Gpr37l1) is expressed by Bergmann astrocytes and interacts with patched 1 (Ptch1) at peri-ciliary membranes. Cerebellar primary astrocyte cultures from wild-type and Gpr37l1 null mutant mouse pups were established and studied. Primary cilia were produced by cultures of both genotypes, as well as Ptch1 and smoothened (Smo) components of the sonic hedgehog (Shh) mitogenic pathway. Compared to wild-type cells, Gpr37l1-/- astrocytes displayed striking increases in proliferative activity, Ptch1 protein expression and internalization, intracellular cholesterol content, ciliary localization of Smo, as well as a marked production of active Shh. Similar effects were reproduced by treating wild-type astrocytes with a putative prosaptide ligand of Gpr37l1. These findings indicate that Gpr37l1-Ptch1 interactions specifically regulate Ptch1 internalization and trafficking, with consequent stimulation of Shh production and activation of proliferative signaling.
Collapse
Affiliation(s)
- Gina La Sala
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome, Italy
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome, Italy
| | - Rafaele Matteoni
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome, Italy
| | - Giulia Bolasco
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory (EMBL), Monterotondo Scalo, Rome, Italy
| | - Daniela Marazziti
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome, Italy
| | - Glauco P Tocchini-Valentini
- Institute of Biochemistry and Cell Biology, Italian National Research Council (CNR), Monterotondo Scalo, Rome, Italy
| |
Collapse
|
48
|
Zhuge W, Zhuge Q, Wang W, Lu X, You R, Liu L, Yu H, Wang J, Wang X, Ye Y, Ding S. Hydrogen sulphide ameliorates dopamine-induced astrocytic inflammation and neurodegeneration in minimal hepatic encephalopathy. J Cell Mol Med 2020; 24:13634-13647. [PMID: 33118312 PMCID: PMC7753993 DOI: 10.1111/jcmm.15728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/19/2022] Open
Abstract
It has been demonstrated that the action of dopamine (DA) could enhance the production of tumour necrosis factor‐α (TNF‐α) by astrocytes and potentiate neuronal apoptosis in minimal hepatic encephalopathy (MHE). Recently, sodium hydrosulfide (NaHS) has been found to have neuroprotective properties. Our study addressed whether NaHS could rescue DA‐challenged inflammation and apoptosis in neurons to ameliorate memory impairment in MHE rats and in the neuron and astrocyte coculture system. We found that NaHS suppressed DA‐induced p65 acetylation, resulting in reduced TNF‐α production in astrocytes both in vitro and in vivo. Furthermore, decreased apoptosis was observed in neurons exposed to conditioned medium from DA + NaHS‐challenged astrocytes, which was similar to the results obtained in the neurons exposed to TNF‐α + NaHS, suggesting a therapeutic effect of NaHS on the suppression of neuronal apoptosis via the reduction of TNF‐α level. DA triggered the inactivation of p70 S6 ribosomal kinase (S6K1) and dephosphorylation of Bad, resulting in the disaggregation of Bclxl and Bak and the release of cytochrome c (Cyt. c), and this process could be reversed by NaHS administration. Our work demonstrated that NaHS attenuated DA‐induced astrocytic TNF‐α release and ameliorated inflammation‐induced neuronal apoptosis in MHE. Further research into this approach may uncover future potential therapeutic strategies for MHE.
Collapse
Affiliation(s)
- Weishan Zhuge
- Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qichuan Zhuge
- Neurosurgery Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weikan Wang
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Xiaoai Lu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruimin You
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Leping Liu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - He Yu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuebao Wang
- Analytical and Testing Center, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiru Ye
- School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Saidan Ding
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
49
|
Meftahi GH, Moafi M, Mirbehbahani SH, Fotouhi F, Toreyhi H, Ezi S, Aghajanpour F, Forouzannia A, Boroujeni ME, Peirouvi T, Abbaszadeh HA, Aliaghaei A. Chronic administration of methylphenidate did not affect memory and GDNF levels but increase astrogliosis in adult male rat’s hippocampus. J Chem Neuroanat 2020; 108:101818. [DOI: 10.1016/j.jchemneu.2020.101818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/02/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022]
|
50
|
Cellular Prion Protein (PrPc): Putative Interacting Partners and Consequences of the Interaction. Int J Mol Sci 2020; 21:ijms21197058. [PMID: 32992764 PMCID: PMC7583789 DOI: 10.3390/ijms21197058] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
Cellular prion protein (PrPc) is a small glycosylphosphatidylinositol (GPI) anchored protein most abundantly found in the outer leaflet of the plasma membrane (PM) in the central nervous system (CNS). PrPc misfolding causes neurodegenerative prion diseases in the CNS. PrPc interacts with a wide range of protein partners because of the intrinsically disordered nature of the protein’s N-terminus. Numerous studies have attempted to decipher the physiological role of the prion protein by searching for proteins which interact with PrPc. Biochemical characteristics and biological functions both appear to be affected by interacting protein partners. The key challenge in identifying a potential interacting partner is to demonstrate that binding to a specific ligand is necessary for cellular physiological function or malfunction. In this review, we have summarized the intracellular and extracellular interacting partners of PrPc and potential consequences of their binding. We also briefly describe prion disease-related mutations at the end of this review.
Collapse
|