51
|
Singh L, Kaur N, Bhatti R. Neuroprotective potential of biochanin-A and review of the molecular mechanisms involved. Mol Biol Rep 2023; 50:5369-5378. [PMID: 37039995 DOI: 10.1007/s11033-023-08397-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/17/2023] [Indexed: 04/12/2023]
Abstract
Biochanin-A is a naturally occurring plant phytoestrogen, which mimics specific the agonistic activity of estrogens. Biochanin-A is known to possess numerous activities, including neuroprotective, anti-diabetic, hepatoprotective, anti-inflammatory, antioxidant, and antimicrobial activities, along with the anticancer activity. Neuroinflammation is thought to play a pivotal pathological role in neurodegenerative disease. Sustained neuroinflammatory processes lead to progressive neuronal damage in Parkinson's and Alzheimer's disease. Activation of PI3K/Akt cascade and inhibition of MAPK signaling cascade have been observed to be responsible for conferring protection against neuroinflammation in neurodegenerative diseases. An increased oxidative stress promotes neuronal apoptosis via potentiating the TLR-4/NF-κB and inhibiting PI3K/Akt signaling mediated increase in pro-apoptotic and decreases in antiapoptotic proteins. Various authors have explored biochanin-A's neuroprotective effect by using various cell lines and animal models. Biochanin-A has been reported to mediate its neuroprotective via reducing the level of oxidants, inflammatory mediators, MAPK, TLR-4, NF-κB, NADPH oxidase, AchE, COX-2 and iNOS. Whereas, it has been observed to increase the level of anti-oxidants, along with phosphorylation of PI3K and Akt proteins. The current review has been designed to provide insights into the neuroprotective effect of biochanin-A and possible signaling pathways leading to protection against neuroinflammation and apoptosis in the central nervous system. This review will be helpful in guiding future researchers to further explore biochanin A at a mechanistic level to obtain useful lead molecules.
Collapse
Affiliation(s)
- Lovedeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India.
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Navneet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Rajbir Bhatti
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
52
|
Isik S, Yeman Kiyak B, Akbayir R, Seyhali R, Arpaci T. Microglia Mediated Neuroinflammation in Parkinson’s Disease. Cells 2023; 12:cells12071012. [PMID: 37048085 PMCID: PMC10093562 DOI: 10.3390/cells12071012] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Parkinson’s Disease (PD) is the second most common neurodegenerative disorder seen, especially in the elderly. Tremor, shaking, movement problems, and difficulty with balance and coordination are among the hallmarks, and dopaminergic neuronal loss in substantia nigra pars compacta of the brain and aggregation of intracellular protein α-synuclein are the pathological characterizations. Neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. It is a complex network of interactions comprising immune and non-immune cells in addition to mediators of the immune response. Microglia, the resident macrophages in the CNS, take on the leading role in regulating neuroinflammation and maintaining homeostasis. Under normal physiological conditions, they exist as “homeostatic” but upon pathological stimuli, they switch to the “reactive state”. Pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes are used to classify microglial activity with each phenotype having its own markers and released mediators. When M1 microglia are persistent, they will contribute to various inflammatory diseases, including neurodegenerative diseases, such as PD. In this review, we focus on the role of microglia mediated neuroinflammation in PD and also signaling pathways, receptors, and mediators involved in the process, presenting the studies that associate microglia-mediated inflammation with PD. A better understanding of this complex network and interactions is important in seeking new therapies for PD and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Uskudar, Istanbul 34662, Turkey
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Correspondence: ; Tel.: +90-216-400-2222 (ext. 2462)
| | - Bercem Yeman Kiyak
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Medicine, Institute of Hamidiye Health Sciences, University of Health Sciences, Uskudar, Istanbul 34668, Turkey
| | - Rumeysa Akbayir
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Rama Seyhali
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Tahire Arpaci
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| |
Collapse
|
53
|
Leng F, Hinz R, Gentleman S, Hampshire A, Dani M, Brooks DJ, Edison P. Neuroinflammation is independently associated with brain network dysfunction in Alzheimer's disease. Mol Psychiatry 2023; 28:1303-1311. [PMID: 36474000 PMCID: PMC10005956 DOI: 10.1038/s41380-022-01878-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/17/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
Brain network dysfunction is increasingly recognised in Alzheimer's disease (AD). However, the causes of brain connectivity disruption are still poorly understood. Recently, neuroinflammation has been identified as an important factor in AD pathogenesis. Microglia participate in the construction and maintenance of healthy neuronal networks, but pro-inflammatory microglia can also damage these circuits. We hypothesised that microglial activation is independently associated with brain connectivity disruption in AD. We performed a cross-sectional multimodal imaging study and interrogated the relationship between imaging biomarkers of neuroinflammation, Aβ deposition, brain connectivity and cognition. 42 participants (12 Aβ-positive MCI, 14 Aβ-positive AD and 16 Aβ-negative healthy controls) were recruited. Participants had 11C-PBR28 and 18F-flutemetamol PET to quantify Aβ deposition and microglial activation, T1-weighted, diffusion tensor and resting-state functional MRI to assess structural network and functional network. 11C-PBR28 uptake, structural network integrity and functional network orgnisation were compared across diagnostic groups and the relationship between neuroinflammation and brain network was tested in 26 Aβ-positive patients. Increased 11C-PBR28 uptake, decreased FA, network small-worldness and local efficiency were observed in AD patients. Cortical 11C-PBR28 uptake correlated negatively with structural integrity (standardised β = -0.375, p = 0.037) and network local efficiency (standardised β = -0.468, p < 0.001), independent of cortical thickness and Aβ deposition, while Aβ was not. Network structural integrity, small-worldness and local efficiency, and cortical thickness were positively associated with cognition. Our findings suggest cortical neuroinflammation coincide with structural and functional network disruption independent of Aβ and cortical atrophy. These findings link the brain connectivity change and pathological process in Alzheimer's disease, and suggest a pathway from neuroinflammation to systemic brain dysfunction.
Collapse
Affiliation(s)
- Fangda Leng
- Department of Brain Sciences, Imperial College London, London, UK
- Department of Neurology, Peking University First Hospital, Beijing, PR China
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Steve Gentleman
- Department of Brain Sciences, Imperial College London, London, UK
| | - Adam Hampshire
- Department of Brain Sciences, Imperial College London, London, UK
| | - Melanie Dani
- Department of Brain Sciences, Imperial College London, London, UK
| | - David J Brooks
- Department of Brain Sciences, Imperial College London, London, UK
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle, UK
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, UK.
- School of Medicine, Cardiff University, Wales, UK.
| |
Collapse
|
54
|
Wu KJ, Wang YS, Hung TW, Bae EK, Chen YH, Kim CK, Yoo DW, Kim GS, Yu SJ. Herbal formula PM012 induces neuroprotection in stroke brain. PLoS One 2023; 18:e0281421. [PMID: 36812289 PMCID: PMC9946208 DOI: 10.1371/journal.pone.0281421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023] Open
Abstract
Stroke is a major cause of long-term disability world-wide. Limited pharmacological therapy has been used in stroke patients. Previous studies indicated that herb formula PM012 is neuroprotective against neurotoxin trimethyltin in rat brain, and improved learning and memory in animal models of Alzheimer's disease. Its action in stroke has not been reported. This study aims to determine PM012-mediated neural protection in cellular and animal models of stroke. Glutamate-mediated neuronal loss and apoptosis were examined in rat primary cortical neuronal cultures. Cultured cells were overexpressed with a Ca++ probe (gCaMP5) by AAV1 and were used to examine Ca++ influx (Ca++i). Adult rats received PM012 before transient middle cerebral artery occlusion (MCAo). Brain tissues were collected for infarction and qRTPCR analysis. In rat primary cortical neuronal cultures, PM012 significantly antagonized glutamate-mediated TUNEL and neuronal loss, as well as NMDA-mediated Ca++i. PM012 significantly reduced brain infarction and improved locomotor activity in stroke rats. PM012 attenuated the expression of IBA1, IL6, and CD86, while upregulated CD206 in the infarcted cortex. ATF6, Bip, CHOP, IRE1, and PERK were significantly down-regulated by PM012. Using HPLC, two potential bioactive molecules, paeoniflorin and 5-hydroxymethylfurfural, were identified in the PM012 extract. Taken together, our data suggest that PM012 is neuroprotective against stroke. The mechanisms of action involve inhibition of Ca++i, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yun-Hsiang Chen
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | | | - Dai-Won Yoo
- Mediforum Co., Ltd., Seoul, Republic of Korea
| | | | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
- * E-mail:
| |
Collapse
|
55
|
Cuní-López C, Stewart R, White AR, Quek H. 3D in vitro modelling of human patient microglia: A focus on clinical translation and drug development in neurodegenerative diseases. J Neuroimmunol 2023; 375:578017. [PMID: 36657374 DOI: 10.1016/j.jneuroim.2023.578017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/28/2022] [Accepted: 01/08/2023] [Indexed: 01/13/2023]
Abstract
Microglia have an increasingly well-recognised role in the pathogenesis of neurodegenerative diseases, thereby becoming attractive therapeutic targets. However, the development of microglia-targeted therapeutics for neurodegeneration has had limited success. This stems partly from the lack of clinically relevant microglia model systems. To circumvent this translational gap, patient-derived microglial cell models established using conventional 2D in vitro techniques have emerged. Though promising, these models lack the microenvironment and multicellular interactions of the brain needed to maintain microglial homeostasis. In this review, we discuss the use of 3D in vitro platforms to improve microglia modelling and their potential benefits to fast-track drug development for neurodegenerative diseases.
Collapse
Affiliation(s)
- Carla Cuní-López
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane 4006, QLD, Australia.
| | - Romal Stewart
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia; UQ Centre for Clinical Research, The University of Queensland, Brisbane 4006, QLD, Australia.
| | - Anthony R White
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia; School of Biomedical Science, The University of Queensland, Brisbane 4072, QLD, Australia.
| | - Hazel Quek
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane 4006, QLD, Australia; School of Biomedical Science, The University of Queensland, Brisbane 4072, QLD, Australia; School of Biomedical Science, Queensland University of Technology, Brisbane 4059, QLD, Australia.
| |
Collapse
|
56
|
Zhao J, Wang X, Li Q, Lu C, Li S. The relevance of serum macrophage migratory inhibitory factor and cognitive dysfunction in patients with cerebral small vascular disease. Front Aging Neurosci 2023; 15:1083818. [PMID: 36824264 PMCID: PMC9941340 DOI: 10.3389/fnagi.2023.1083818] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Cerebral small vascular disease (CSVD) is a common type of cerebrovascular disease, and an important cause of vascular cognitive impairment (VCI) and stroke. The disease burden is expected to increase further as a result of population aging, an ongoing high prevalence of risk factors (e.g., hypertension), and inadequate management. Due to the poor understanding of pathophysiology in CSVD, there is no effective preventive or therapeutic approach for CSVD. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine that is related to the occurrence and development of vascular dysfunction diseases. Therefore, MIF may contribute to the pathogenesis of CSVD and VCI. Here, reviewed MIF participation in chronic cerebral ischemia-hypoperfusion and neurodegeneration pathology, including new evidence for CSVD, and its potential role in protection against VCI.
Collapse
Affiliation(s)
- Jianhua Zhao
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China,*Correspondence: Jianhua Zhao,
| | - Xiaoting Wang
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qiong Li
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Chengbiao Lu
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
57
|
Pinosanu LR, Capitanescu B, Glavan D, Godeanu S, Cadenas IF, Doeppner TR, Hermann DM, Balseanu AT, Bogdan C, Popa-Wagner A. Neuroglia Cells Transcriptomic in Brain Development, Aging and Neurodegenerative Diseases. Aging Dis 2023; 14:63-83. [PMID: 36818562 PMCID: PMC9937697 DOI: 10.14336/ad.2022.0621] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Glia cells are essential for brain functioning during development, aging and disease. However, the role of astroglia plays during brain development is quite different from the role played in the adult lesioned brain. Therefore, a deeper understanding of pathomechanisms underlying astroglia activity in the aging brain and cerebrovascular diseases is essential to guide the development of new therapeutic strategies. To this end, this review provides a comparison between the transcriptomic activity of astroglia cells during development, aging and neurodegenerative diseases, including cerebral ischemia. During fetal brain development, astrocytes and microglia often affect the same developmental processes such as neuro-/gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis, and synaptic pruning. In the adult brain astrocytes are a critical player in the synapse remodeling by mediating synapse elimination while microglia activity has been associated with changes in synaptic plasticity and remove cell debris by constantly sensing the environment. However, in the lesioned brain astrocytes proliferate and play essential functions with regard to energy supply to the neurons, neurotransmission and buildup of a protective scar isolating the lesion site from the surroundings. Inflammation, neurodegeneration, or loss of brain homeostasis induce changes in microglia gene expression, morphology, and function, generally referred to as "primed" microglia. These changes in gene expression are characterized by an enrichment of phagosome, lysosome, and antigen presentation signaling pathways and is associated with an up-regulation of genes encoding cell surface receptors. In addition, primed microglia are characterized by upregulation of a network of genes in response to interferon gamma. Conclusion. A comparison of astroglia cells transcriptomic activity during brain development, aging and neurodegenerative disorders might provide us with new therapeutic strategies with which to protect the aging brain and improve clinical outcome.
Collapse
Affiliation(s)
- Leonard Radu Pinosanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Bogdan Capitanescu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Daniela Glavan
- Psychiatric clinic, University of Medicine and Pharmacy Craiova, Craiova, Romania.
| | - Sanziana Godeanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Israel Ferna´ndez Cadenas
- Stroke Pharmacogenomics and Genetics group, Sant Pau Hospital Institute of Research, Barcelona, Spain.
| | - Thorsten R. Doeppner
- Department of Neurology, University Hospital Giessen, Giessen, Germany.,University of Göttingen Medical School, Department of Neurology, Göttingen, Germany.
| | - Dirk M. Hermann
- Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.
| | - Adrian-Tudor Balseanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Catalin Bogdan
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.,Correspondence should be addressed to: Dr. Aurel Popa-Wagner () and Dr. Catalin Bogdan (), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.,Correspondence should be addressed to: Dr. Aurel Popa-Wagner () and Dr. Catalin Bogdan (), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| |
Collapse
|
58
|
Anwar MM, Fathi MH. Early approaches of YKL-40 as a biomarker and therapeutic target for Parkinson's disease. Neurodegener Dis Manag 2023; 13:85-99. [PMID: 36644988 DOI: 10.2217/nmt-2022-0010] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aim: To investigate whether the estimation of cerebrospinal fluid (CSF) and brain YKL-40 levels may be used as an efficient biomarker for Parkinson's disease (PD). Methods: Lipopolysaccharides (LPS) was injected into the right substantia nigra pars compacta (SNpc). Rats were divided into: control group, early LPS-induced PD group (14 days), and advanced LPS-induced PD group (28 days). YKL-40 and other related factors were detected in CSF and brain tissue. Results: Increased expression of YKL-40 was observed in brain tissue and CSF of PD-induced rats associated with triggered inflammatory cytokine release. Conclusion: The current study was limited to detecting YKL-40 and other inflammatory factors in brain and CSF. YKL-40 may be considered as an early biomarker and therapeutic target for PD.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control & Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Mohamed H Fathi
- Department of Nucleic Acid & Protein structure, Center of Genomics, Proteomics & Bioinformatics, Agricultural Genetic Engineering Research Institute (AGERI), Cairo, Egypt
| |
Collapse
|
59
|
de Almeida MMA, Goodkey K, Voronova A. Regulation of microglia function by neural stem cells. Front Cell Neurosci 2023; 17:1130205. [PMID: 36937181 PMCID: PMC10014810 DOI: 10.3389/fncel.2023.1130205] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Neural stem and precursor cells (NPCs) build and regenerate the central nervous system (CNS) by maintaining their pool (self-renewal) and differentiating into neurons, astrocytes, and oligodendrocytes (multipotency) throughout life. This has inspired research into pro-regenerative therapies that utilize transplantation of exogenous NPCs or recruitment of endogenous adult NPCs for CNS regeneration and repair. Recent advances in single-cell RNA sequencing and other "omics" have revealed that NPCs express not just traditional progenitor-related genes, but also genes involved in immune function. Here, we review how NPCs exert immunomodulatory function by regulating the biology of microglia, immune cells that are present in NPC niches and throughout the CNS. We discuss the role of transplanted and endogenous NPCs in regulating microglia fates, such as survival, proliferation, migration, phagocytosis and activation, in the developing, injured and degenerating CNS. We also provide a literature review on NPC-specific mediators that are responsible for modulating microglia biology. Our review highlights the immunomodulatory properties of NPCs and the significance of these findings in the context of designing pro-regenerative therapies for degenerating and diseased CNS.
Collapse
Affiliation(s)
- Monique M. A. de Almeida
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, Canada
| | - Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, Canada
- Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
| |
Collapse
|
60
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
61
|
TDP-43 condensates and lipid droplets regulate the reactivity of microglia and regeneration after traumatic brain injury. Nat Neurosci 2022; 25:1608-1625. [PMID: 36424432 DOI: 10.1038/s41593-022-01199-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/11/2022] [Indexed: 11/27/2022]
Abstract
Decreasing the activation of pathology-activated microglia is crucial to prevent chronic inflammation and tissue scarring. In this study, we used a stab wound injury model in zebrafish and identified an injury-induced microglial state characterized by the accumulation of lipid droplets and TAR DNA-binding protein of 43 kDa (TDP-43)+ condensates. Granulin-mediated clearance of both lipid droplets and TDP-43+ condensates was necessary and sufficient to promote the return of microglia back to the basal state and achieve scarless regeneration. Moreover, in postmortem cortical brain tissues from patients with traumatic brain injury, the extent of microglial activation correlated with the accumulation of lipid droplets and TDP-43+ condensates. Together, our results reveal a mechanism required for restoring microglia to a nonactivated state after injury, which has potential for new therapeutic applications in humans.
Collapse
|
62
|
Immunosenescence and Aging: Neuroinflammation Is a Prominent Feature of Alzheimer's Disease and Is a Likely Contributor to Neurodegenerative Disease Pathogenesis. J Pers Med 2022; 12:jpm12111817. [PMID: 36579548 PMCID: PMC9698256 DOI: 10.3390/jpm12111817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic multifactorial and complex neuro-degenerative disorder characterized by memory impairment and the loss of cognitive ability, which is a problem affecting the elderly. The pathological intracellular accumulation of abnormally phosphorylated Tau proteins, forming neurofibrillary tangles, and extracellular amyloid-beta (Aβ) deposition, forming senile plaques, as well as neural disconnection, neural death and synaptic dysfunction in the brain, are hallmark pathologies that characterize AD. The prevalence of the disease continues to increase globally due to the increase in longevity, quality of life, and medical treatment for chronic diseases that decreases the mortality and enhance the survival of elderly. Medical awareness and the accurate diagnosis of the disease also contribute to the high prevalence observed globally. Unfortunately, no definitive treatment exists that can be used to modify the course of AD, and no available treatment is capable of mitigating the cognitive decline or reversing the pathology of the disease as of yet. A plethora of hypotheses, ranging from the cholinergic theory and dominant Aβ cascade hypothesis to the abnormally excessive phosphorylated Tau protein hypothesis, have been reported. Various explanations for the pathogenesis of AD, such as the abnormal excitation of the glutamate system and mitochondrial dysfunction, have also been suggested. Despite the continuous efforts to deliver significant benefits and an effective treatment for this distressing, globally attested aging illness, multipronged approaches and strategies for ameliorating the disease course based on knowledge of the underpinnings of the pathogenesis of AD are urgently needed. Immunosenescence is an immune deficit process that appears with age (inflammaging process) and encompasses the remodeling of the lymphoid organs, leading to alterations in the immune function and neuroinflammation during advanced aging, which is closely linked to the outgrowth of infections, autoimmune diseases, and malignant cancers. It is well known that long-standing inflammation negatively influences the brain over the course of a lifetime due to the senescence of the immune system. Herein, we aim to trace the role of the immune system in the pathogenesis of AD. Thus, we explore alternative avenues, such as neuroimmune involvement in the pathogenesis of AD. We determine the initial triggers of neuroinflammation, which is an early episode in the pre-symptomatic stages of AD and contributes to the advancement of the disease, and the underlying key mechanisms of brain damage that might aid in the development of therapeutic strategies that can be used to combat this devastating disease. In addition, we aim to outline the ways in which different aspects of the immune system, both in the brain and peripherally, behave and thus to contribute to AD.
Collapse
|
63
|
Zhu R, Luo Y, Li S, Wang Z. The role of microglial autophagy in Parkinson's disease. Front Aging Neurosci 2022; 14:1039780. [PMID: 36389074 PMCID: PMC9664157 DOI: 10.3389/fnagi.2022.1039780] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/10/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Studies have shown that abnormal accumulation of α-synuclein (α-Syn) in the substantia nigra is a specific pathological characteristic of PD. Abnormal accumulation of α-Syn in PD induces the activation of microglia. Microglia, which are immune cells in the central nervous system, are involved in the function and regulation of inflammation in PD by autophagy. The role of microglial autophagy in the pathophysiology of PD has become a hot-pot issue. This review outlines the pathways of microglial autophagy, and explores the key factor of microglial autophagy in the mechanism of PD and the possibility of microglial autophagy as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Rui Zhu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yuyi Luo
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China,*Correspondence: Zhengbo Wang,
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China,*Correspondence: Zhengbo Wang,
| |
Collapse
|
64
|
An anti-inflammatory transcriptional cascade conserved from flies to humans. Cell Rep 2022; 41:111506. [DOI: 10.1016/j.celrep.2022.111506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
|
65
|
Advanced therapeutic strategies targeting microglia: beyond neuroinflammation. Arch Pharm Res 2022; 45:618-630. [PMID: 36166145 DOI: 10.1007/s12272-022-01406-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
For a long time, microglia have been recognized as the main culprits of neuroinflammatory responses because they are primary phagocytes present in the parenchyma of the central nervous system (CNS). However, with the evolving concept of microglial biology, advanced and precise approaches, rather than the global inhibition of activated microglia, have been proposed in the management of neurological disorders. Yolk sac-derived resident microglia have heterogeneous composition according to brain region, sex, and diseases. They play a key role in the maintenance of CNS homeostasis and as primary phagocytes. The perturbation of microglia development can induce neurodevelopmental disorders. Microglia aggravate or alleviate neuroinflammation according to microenvironment and their spatiotemporal dynamics. They are long-lived cells and repopulate via their proliferation or external monocyte engraft. Based on this evolving concept, understanding advanced therapeutic strategies targeting microglia can give us an opportunity to discover novel therapies for neurological disorders.
Collapse
|
66
|
Mo Y, Xu W, Fu K, Chen H, Wen J, Huang Q, Guo F, Mo L, Yan J. The dual function of microglial polarization and its treatment targets in ischemic stroke. Front Neurol 2022; 13:921705. [PMID: 36212660 PMCID: PMC9538667 DOI: 10.3389/fneur.2022.921705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Stroke is the leading cause of disability and death worldwide, with ischemic stroke occurring in ~5% of the global population every year. Recently, many studies have been conducted on the inflammatory response after stroke. Microglial/macrophage polarization has a dual function and is critical to the pathology of ischemic stroke. Microglial/macrophage activation is important in reducing neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after ischemic stroke. In this review, we investigate the physiological characteristics and functions of microglia in the brain, the activation and phenotypic polarization of microglia and macrophages after stroke, the signaling mechanisms of polarization states, and the contribution of microglia to brain pathology and repair. We summarize recent advances in stroke-related microglia research, highlighting breakthroughs in therapeutic strategies for microglial responses after stroke, thereby providing new ideas for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yong Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kaijing Fu
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hainan Chen
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jing Wen
- Department of Rheumatism, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qianrong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fangzhou Guo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Ligen Mo
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Jun Yan
| |
Collapse
|
67
|
Ferroptosis is involved in regulating perioperative neurocognitive disorders: emerging perspectives. J Neuroinflammation 2022; 19:219. [PMID: 36068571 PMCID: PMC9450301 DOI: 10.1186/s12974-022-02570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
Since the twenty-first century, the development of technological advances in anesthesia and surgery has brought benefits to human health. However, the adverse neurological effects of perioperative-related factors (e.g., surgical trauma, anesthesia, etc.) as stressors cannot be ignored as well. The nervous system appears to be more "fragile" and vulnerable to damage in developing and aging individuals. Ferroptosis is a novel form of programmed cell death proposed in 2012. In recent years, the regulation of ferroptosis to treat cancer, immune system disorders, and neurodegenerative diseases have seen an unprecedented surge of interest. The association of ferroptosis with perioperative neurocognitive disorders has also received much attention. Cognitive impairment can not only affect the individual's quality of life, but also impose a burden on the family and society. Therefore, the search for effective preventive and therapeutic methods to alleviate cognitive impairment caused by perioperative-related factors is a challenge that needs to be urgently addressed. In our review, we first briefly describe the connection between iron accumulation in neurons and impairment of brain function during development and aging. It is followed by a review of the pathways of ferroptosis, mainly including iron metabolism, amino acid metabolism, and lipid metabolism pathway. Furthermore, we analyze the connection between ferroptosis and perioperative-related factors. The surgery itself, general anesthetic drugs, and many other relevant factors in the perioperative period may affect neuronal iron homeostasis. Finally, we summarize the experimental evidence for ameliorating developmental and degenerative neurotoxicity by modulating ferroptosis. The suppression of ferroptosis seems to provide the possibility to prevent and improve perioperative neurocognitive impairment.
Collapse
|
68
|
Errede M, Annese T, Petrosino V, Longo G, Girolamo F, de Trizio I, d'Amati A, Uccelli A, Kerlero de Rosbo N, Virgintino D. Microglia-derived CCL2 has a prime role in neocortex neuroinflammation. Fluids Barriers CNS 2022; 19:68. [PMID: 36042496 PMCID: PMC9429625 DOI: 10.1186/s12987-022-00365-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/03/2022] [Indexed: 11/12/2022] Open
Abstract
Background In myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), several areas of demyelination are detectable in mouse cerebral cortex, where neuroinflammation events are associated with scarce inflammatory infiltrates and blood–brain barrier (BBB) impairment. In this condition, the administration of mesenchymal stem cells (MSCs) controls neuroinflammation, attenuating astrogliosis and promoting the acquisition of stem cell traits by astrocytes. To contribute to the understanding of the mechanisms involved in the pathogenesis of EAE in gray matter and in the reverting effects of MSC treatment, the neocortex of EAE-affected mice was investigated by analyzing the cellular source(s) of chemokine CCL2, a molecule involved in immune cell recruitment and BBB-microvessel leakage. Methods The study was carried out by immunohistochemistry (IHC) and dual RNAscope IHC/in situ hybridization methods, using astrocyte, NG2-glia, macrophage/microglia, and microglia elective markers combined with CCL2. Results The results showed that in EAE-affected mice, hypertrophic microglia are the primary source of CCL2, surround the cortex neurons and the damaged BBB microvessels. In EAE-affected mice treated with MSCs, microgliosis appeared diminished very soon (6 h) after treatment, an observation that was long-lasting (tested after 10 days). This was associated with a reduced CCL2 expression and with apparently preserved/restored BBB features. In conclusion, the hallmark of EAE in the mouse neocortex is a condition of microgliosis characterized by high levels of CCL2 expression. Conclusions This finding supports relevant pathogenetic and clinical aspects of the human disease, while the demonstrated early control of neuroinflammation and BBB permeability exerted by treatment with MSCs may have important therapeutic implications. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00365-5.
Collapse
Affiliation(s)
- Mariella Errede
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.,Department of Medicine and Surgery, LUM University, Casamassima Bari, Italy
| | - Valentina Petrosino
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicole Kerlero de Rosbo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,TomaLab, Institute of Nanotechnology, Consiglio Nazionale Delle Ricerche (CNR), Rome, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.
| |
Collapse
|
69
|
Zhao F, Li B, Yang W, Ge T, Cui R. Brain-immune interaction mechanisms: Implications for cognitive dysfunction in psychiatric disorders. Cell Prolif 2022; 55:e13295. [PMID: 35860850 PMCID: PMC9528770 DOI: 10.1111/cpr.13295] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/28/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
Objectives Cognitive dysfunction has been identified as a major symptom of a series of psychiatric disorders. Multidisciplinary studies have shown that cognitive dysfunction is monitored by a two‐way interaction between the neural and immune systems. However, the specific mechanisms of cognitive dysfunction in immune response and brain immune remain unclear. Materials and methods In this review, we summarized the relevant research to uncover our comprehension of the brain–immune interaction mechanisms underlying cognitive decline. Results The pathophysiological mechanisms of brain‐immune interactions in psychiatric‐based cognitive dysfunction involve several specific immune molecules and their associated signaling pathways, impairments in neural and synaptic plasticity, and the potential neuro‐immunological mechanism of stress. Conclusions Therefore, this review may provide a better theoretical basis for integrative therapeutic considerations for psychiatric disorders associated with cognitive dysfunction.
Collapse
Affiliation(s)
- Fangyi Zhao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
70
|
Choi J, Kim BR, Akuzum B, Chang L, Lee JY, Kwon HK. TREGking From Gut to Brain: The Control of Regulatory T Cells Along the Gut-Brain Axis. Front Immunol 2022; 13:916066. [PMID: 35844606 PMCID: PMC9279871 DOI: 10.3389/fimmu.2022.916066] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
The human gastrointestinal tract has an enormous and diverse microbial community, termed microbiota, that is necessary for the development of the immune system and tissue homeostasis. In contrast, microbial dysbiosis is associated with various inflammatory and autoimmune diseases as well as neurological disorders in humans by affecting not only the immune system in the gastrointestinal tract but also other distal organs. FOXP3+ regulatory T cells (Tregs) are a subset of CD4+ helper T cell lineages that function as a gatekeeper for immune activation and are essential for peripheral autoimmunity prevention. Tregs are crucial to the maintenance of immunological homeostasis and tolerance at barrier regions. Tregs reside in both lymphoid and non-lymphoid tissues, and tissue-resident Tregs have unique tissue-specific phenotype and distinct function. The gut microbiota has an impact on Tregs development, accumulation, and function in periphery. Tregs, in turn, modulate antigen-specific responses aimed towards gut microbes, which supports the host–microbiota symbiotic interaction in the gut. Recent studies have indicated that Tregs interact with a variety of resident cells in central nervous system (CNS) to limit the progression of neurological illnesses such as ischemic stroke, Alzheimer’s disease, and Parkinson’s disease. The gastrointestinal tract and CNS are functionally connected, and current findings provide insights that Tregs function along the gut-brain axis by interacting with immune, epithelial, and neuronal cells. The purpose of this study is to explain our current knowledge of the biological role of tissue-resident Tregs, as well as the interaction along the gut-brain axis.
Collapse
Affiliation(s)
- Juli Choi
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Bo-Ram Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
| | - Begum Akuzum
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
| | - Leechung Chang
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: June-Yong Lee, ; Ho-Keun Kwon,
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, South Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: June-Yong Lee, ; Ho-Keun Kwon,
| |
Collapse
|
71
|
Fu S, Zhao S, Chen H, Yang W, Xia X, Xu X, Liang Z, Feng X, Wang Z, Ai P, Ding L, Cai Q, Wang Y, Zhang Y, Zhu J, Zhang B, Zheng JC. Insulin-incubated palladium clusters promote recovery after brain injury. J Nanobiotechnology 2022; 20:299. [PMID: 35752849 PMCID: PMC9233827 DOI: 10.1186/s12951-022-01495-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/04/2022] [Indexed: 11/10/2022] Open
Abstract
Traumatic brain injury (TBI) is a cause of disability and death worldwide, but there are currently no specific treatments for this condition. Release of excess reactive oxygen species (ROS) in the injured brain leads to a series of pathological changes; thus, eliminating ROS could be a potential therapeutic strategy. Herein, we synthesized insulin-incubated ultrasmall palladium (Pd@insulin) clusters via green biomimetic chemistry. The Pd@insulin clusters, which were 3.2 nm in diameter, exhibited marked multiple ROS-scavenging ability testified by the theoretical calculation. Pd@insulin could be rapidly excreted via kidney-urine metabolism and induce negligible adverse effects after a long-time treatment in vivo. In a TBI mouse model, intravenously injected Pd@insulin clusters aggregated in the injured cortex, effectively suppressed excessive ROS production, and significantly rescued motor function, cognition and spatial memory. We found that the positive therapeutic effects of the Pd@insulin clusters were mainly attributed to their ROS-scavenging ability, as they inhibited excessive neuroinflammation, reduced cell apoptosis, and prevented neuronal loss. Therefore, the ability of Pd@insulin clusters to effectively eliminate ROS, as well as their simple structure, easy synthesis, low toxicity, and rapid metabolism may facilitate their clinical translation for TBI treatment.
Collapse
Affiliation(s)
- Shengyang Fu
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Shu Zhao
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huili Chen
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Weitao Yang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, Shanghai, 200120, China.,The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China.,Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University School of Medicine, Shanghai, 200331, China
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China. .,Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University School of Medicine, Shanghai, 200331, China. .,Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200434, China. .,Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai, 200065, China.
| | - Xiaonan Xu
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zhanping Liang
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xuanran Feng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zhuo Wang
- The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Pu Ai
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.,Wuxi Clinical College of Anhui Medical University, Hefei, 230022, China
| | - Lu Ding
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Qingyuan Cai
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.,Franklin & Marshall College, Lancaster, PA, 17603, United States
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Yangzhi Rehabilitation Hospital affiliated to Tongji University, Shanghai, 200065, China
| | - Yanyan Zhang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jie Zhu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Bingbo Zhang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, Shanghai, 200120, China. .,The Institute for Biomedical Engineering & Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China. .,Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University School of Medicine, Shanghai, 200331, China.
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China. .,Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University School of Medicine, Shanghai, 200331, China. .,Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200434, China. .,Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai, 200065, China.
| |
Collapse
|
72
|
Bianchin MM, Snow Z. Primary microglia dysfunction or microgliopathy: A cause of dementias and other neurological or psychiatric disorders. Neuroscience 2022; 497:324-339. [PMID: 35760218 DOI: 10.1016/j.neuroscience.2022.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022]
Abstract
Microglia are unique cells in the central nervous system (CNS), being considered a sub-type of CNS macrophage. These cells monitor nearby micro-regions, having roles that far exceed immunological and scavengering functions, being fundamental for developing, protecting and maintaining the integrity of grey and white matter. Microglia might become dysfunctional, causing abnormal CNS functioning early or late in the life of patients, leading to neurologic or psychiatric disorders and premature death in some patients. Observations that the impairment of normal microglia function per se could lead to neurological or psychiatric diseases have been mainly obtained from genetic and molecular studies of Nasu-Hakola disease, caused by TYROBP or TREM2 mutations, and from studies of adult-onset leukoencephalopathy with axonal spheroids (ALSP), caused by CSF1R mutations. These classical microgliopathies are being named here Microgliopathy Type I. Recently, mutations in TREM2 have also been associated with Alzheimer Disease. However, in Alzheimer Disease TREM2 allele variants lead to an impaired, but functional TREM2 protein, so that patients do not develop Nasu-Hakola disease but are at increased risk to develop other neurodegenerative diseases. Alzheimer Disease is the prototype of the neurodegenerative disorders associated with these TREM2 variants, named here the Microgliopathies Type II. Here, we review clinical, pathological and some molecular aspects of human diseases associated with primary microglia dysfunctions and briefly comment some possible therapeutic approaches to theses microgliopathies. We hope that our review might update the interesting discussion about the impact of intrinsic microglia dysfunctions in the genesis of some pathologic processes of the CNS.
Collapse
Affiliation(s)
- Marino Muxfeldt Bianchin
- Basic Research and Advanced Investigations in Neurosciences (BRAIN), Universidade Federal do Rio Grande do Sul, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Brazil; Centro de Tratamento de Epilepsia Refratária (CETER), Hospital de Clínicas de Porto Alegre, Brazil; Division of Neurology, Hospital de Clínicas de Porto Alegre, Brazil.
| | - Zhezu Snow
- Basic Research and Advanced Investigations in Neurosciences (BRAIN), Universidade Federal do Rio Grande do Sul, Brazil
| |
Collapse
|
73
|
Grotemeyer A, McFleder RL, Wu J, Wischhusen J, Ip CW. Neuroinflammation in Parkinson's Disease - Putative Pathomechanisms and Targets for Disease-Modification. Front Immunol 2022; 13:878771. [PMID: 35663989 PMCID: PMC9158130 DOI: 10.3389/fimmu.2022.878771] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive and debilitating chronic disease that affects more than six million people worldwide, with rising prevalence. The hallmarks of PD are motor deficits, the spreading of pathological α-synuclein clusters in the central nervous system, and neuroinflammatory processes. PD is treated symptomatically, as no causally-acting drug or procedure has been successfully established for clinical use. Various pathways contributing to dopaminergic neuron loss in PD have been investigated and described to interact with the innate and adaptive immune system. We discuss the possible contribution of interconnected pathways related to the immune response, focusing on the pathophysiology and neurodegeneration of PD. In addition, we provide an overview of clinical trials targeting neuroinflammation in PD.
Collapse
Affiliation(s)
| | | | - Jingjing Wu
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Jörg Wischhusen
- Section for Experimental Tumor Immunology, Department of Obstetrics and Gynecology, University Hospital of Würzburg, Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
74
|
Ye C, Cheng M, Ma L, Zhang T, Sun Z, Yu C, Wang J, Dou Y. Oxytocin Nanogels Inhibit Innate Inflammatory Response for Early Intervention in Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21822-21835. [PMID: 35510352 DOI: 10.1021/acsami.2c00007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Prevention of Alzheimer's disease (AD) is a global imperative, but reliable early interventions are currently lacking. Microglia-mediated chronic neuroinflammation is thought to occur in the early stage of AD and plays a critical role in AD pathogenesis. Here, oxytocin (OT)-loaded angiopep-2-modified chitosan nanogels (AOC NGs) were designed for early treatment of AD via inhibiting innate inflammatory response. Through the effective transcytosis of angiopep-2, AOC NGs were driven intravenously to cross the blood-brain barrier, enter the brain, and enrich in brain areas affected by AD. A large amount of OT was then released and specifically bound to the pathological upregulated OT receptor, thus effectively inhibiting microglial activation and reducing inflammatory cytokine levels through blocking the ERK/p38 MAPK and COX-2/iNOS NF-κB signaling pathways. Consecutive weekly intravenous administration of AOC NGs into 12-week-old young APP/PS1 mice, representing the early stage of AD, remarkably slowed the progression of Aβ deposition and neuronal apoptosis in the APP/PS1 mice as they aged and ultimately prevented cognitive impairment and delayed hippocampal atrophy. Together, the findings suggest that AOC NGs, which show good biosafety, can serve as a promising therapeutic candidate to combat neuroinflammation for early prevention of AD.
Collapse
Affiliation(s)
- Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Lin Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| |
Collapse
|
75
|
Dai Y, Yan M, Wan J, Xiao T. Maf1 mitigates sevoflurane-induced microglial inflammatory damage and attenuates microglia-mediated neurotoxicity in HT-22 cells by activating the AMPK/Nrf2 signaling. Neurotoxicology 2022; 90:237-245. [PMID: 35430185 DOI: 10.1016/j.neuro.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/26/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Maf1 has been found to play protective function against neuroinflammation and neuroapoptosis. This study seeks to explore whether and how Maf1 is involved in sevoflurane (Sev)-induced neuroinflammation and microglia-mediated neurotoxicity. METHODS qRT-PCR and western blot were used to detect the gene expression. ELISA was used to detect inflammatory factors. Cell viability was evaluated by using the Cell Counting Kit-8 kit. Neuroapoptosis was assessed with trhe Caspase-3 Assay Kit and the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) technique. RESULTS Maf1 expression was downregulated in Sev-stimulated BV2 microglial cells. Maf1 overexpression down-regulates the expression of pro-inflammatory M1-type markers (CD86, iNOS, IFN-γ) and up-regulates the expression of anti-inflammatory M2-type markers (CD206, TGF-β, Arg-1), and Maf1 reduces the Sev-induced inflammatory response in BV2 cells. After Maf1 overexpression, the relative expression of p-AMPK/AMPK and nucleus-Nrf2 increased significantly in BV2 cells treated with Sev. Inhibition of AMPK/Nrf2 pathway by compound C reverses anti-inflammatory effect of Maf1 in Sev-stimulated BV2 cells. Compound C reverses the effect of Maf1 on microglia-mediated neurotoxicity in HT-22 hippocampal neuronal cells. CONCLUSIONS Maf1 mitigates Sev-induced microglial inflammatory damage and attenuates microglia-mediated neurotoxicity by activating the AMPK/Nrf2 signaling.
Collapse
Affiliation(s)
- Yunyi Dai
- Department of Neurology, The First People's Hospital of Shangqiu, China
| | - Mingguang Yan
- Department of Laboratory Medicine, The First People's Hospital of Shangqiu, China
| | - Juan Wan
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Tao Xiao
- Department of Neurosurgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, China.
| |
Collapse
|
76
|
The Impact of Mouse Preterm Birth Induction by RU-486 on Microglial Activation and Subsequent Hypomyelination. Int J Mol Sci 2022; 23:ijms23094867. [PMID: 35563258 PMCID: PMC9105222 DOI: 10.3390/ijms23094867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Preterm birth (PTB) represents 15 million births every year worldwide and is frequently associated with maternal/fetal infections and inflammation, inducing neuroinflammation. This neuroinflammation is mediated by microglial cells, which are brain-resident macrophages that release cytotoxic molecules that block oligodendrocyte differentiation, leading to hypomyelination. Some preterm survivors can face lifetime motor and/or cognitive disabilities linked to periventricular white matter injuries (PWMIs). There is currently no recommendation concerning the mode of delivery in the case of PTB and its impact on brain development. Many animal models of induced-PTB based on LPS injections exist, but with a low survival rate. There is a lack of information regarding clinically used pharmacological substances to induce PTB and their consequences on brain development. Mifepristone (RU-486) is a drug used clinically to induce preterm labor. This study aims to elaborate and characterize a new model of induced-PTB and PWMIs by the gestational injection of RU-486 and the perinatal injection of pups with IL-1beta. A RU-486 single subcutaneous (s.c.) injection at embryonic day (E)18.5 induced PTB at E19.5 in pregnant OF1 mice. All pups were born alive and were adopted directly after birth. IL-1beta was injected intraperitoneally from postnatal day (P)1 to P5. Animals exposed to both RU-486 and IL-1beta demonstrated microglial reactivity and subsequent PWMIs. In conclusion, the s.c. administration of RU-486 induced labor within 24 h with a high survival rate for pups. In the context of perinatal inflammation, RU-486 labor induction significantly decreases microglial reactivity in vivo but did not prevent subsequent PWMIs.
Collapse
|
77
|
Jiang M, Huang Y, Hu L, Wu H, Liu Y, Ni K, Zhang X, Sun Y, Gu X. The transcription factor CCAAT/enhancer-binding protein β in spinal microglia contributes to pre-operative stress-induced prolongation of postsurgical pain. Mol Pain 2022; 18:17448069221099360. [PMID: 35451875 PMCID: PMC9257637 DOI: 10.1177/17448069221099360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Prolongation of postsurgical pain caused by pre-operative stress is a clinically significant problem, although the mechanisms are not fully understood. Stress can promote the pro-inflammatory activation of microglia, and the transcription factor CCAAT/enhancer-binding protein (C/EBP) β regulates pro-inflammatory gene expression in microglia. Therefore, we speculated that C/EBPβ in spinal microglia may have critical roles in the development of chronic postsurgical pain. Accordingly, in this study, we used a single prolonged stress (SPS) procedure and plantar incisions to evaluate the roles of C/EBPβ in postsurgical pain. Our experiments showed that SPS exposure prolonged mechanical allodynia, increased the expression of C/EBPβ and pro-inflammatory cytokines, and potentiated the activation of spinal microglia. Subsequently, microinjection of C/EBPβ siRNA attenuated the duration of SPS-prolonged postoperative mechanical allodynia and inhibited microglial activation in the spinal cord. Conversely, mimicking this increase in C/EBPβ promoted microglial activation via pretreatment with a pre-injection of AAV5-C/EBPβ, leading to prolongation of postsurgical pain. Overall, these results suggested that spinal microglia may play key roles in prolongation of postsurgical pain induced by pre-operative stress and that C/EBPβ may be a potential target for disease treatment.
Collapse
Affiliation(s)
- Ming Jiang
- Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Yulin Huang
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Lijun Hu
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Hao Wu
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Yue Liu
- Department of Anesthesiology66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Kun Ni
- Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Xiaokun Zhang
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Yu'e Sun
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Xiaoping Gu
- Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| |
Collapse
|
78
|
Maurya SK, Mishra R. Molecular docking studies of natural immunomodulators indicate their interactions with the CD40L of CD40/CD40L pathway and CSF1R kinase domain of microglia. J Mol Model 2022; 28:101. [PMID: 35325302 DOI: 10.1007/s00894-022-05084-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
Abstract
Natural products have proved beneficial in reducing neuroinflammation in neurological diseases. Their impacts have also been associated with the activities of microglia, responsible for brain-specific immunity. Recent studies have shown the involvement of the number of microglia-specific proteins in the regulation of brain-specific immunity. However, molecular targets of natural products and their mechanism of interaction with microglia-specific proteins are elusive. Since the genetic signature of microglia offers many potential targets for drug discovery, molecular docking followed by molecular dynamics (MD) simulations of cluster of differentiation 40 ligand (CD40L) and colony-stimulating factor 1 receptor (CSF1R) kinase domain protein with some known neuro-immunomodulators (Curcumin, Cannabidiol, Ginsenoside Rg1, Resveratrol, and Sulforaphane) has been evaluated. Curcumin and cannabidiol were observed likely to modulate CD40L and expression of cytokines and entry of inflammatory cells. Resveratrol and cannabidiol may affect the CSF1R kinase domain and activation of microglia. Our finding suggests that curcumin, cannabidiol, and resveratrol may serve specific drug ligands in regulating microglia-mediated brain immunity.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Department of Zoology, Ramjas College, University of Delhi, 110007, Delhi, India.,Department of Zoology, School of Sciences, Cluster University of Jammu, 180001, Jammu, India.,Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, 221005, Varanasi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, 221005, Varanasi, India.
| |
Collapse
|
79
|
Aguilar-Castillo MJ, Cabezudo-García P, Ciano-Petersen NL, García-Martin G, Marín-Gracia M, Estivill-Torrús G, Serrano-Castro PJ. Immune Mechanism of Epileptogenesis and Related Therapeutic Strategies. Biomedicines 2022; 10:716. [PMID: 35327518 PMCID: PMC8945207 DOI: 10.3390/biomedicines10030716] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/05/2023] Open
Abstract
Immunologic and neuroinflammatory pathways have been found to play a major role in the pathogenesis of many neurological disorders such as epilepsy, proposing the use of novel therapeutic strategies. In the era of personalized medicine and in the face of the exhaustion of anti-seizure therapeutic resources, it is worth looking at the current or future possibilities that neuroimmunomodulator or anti-inflammatory therapy can offer us in the management of patients with epilepsy. For this reason, we performed a narrative review on the recent advances on the basic epileptogenic mechanisms related to the activation of immunity or neuroinflammation with special attention to current and future opportunities for novel treatments in epilepsy. Neuroinflammation can be considered a universal phenomenon and occurs in structural, infectious, post-traumatic, autoimmune, or even genetically based epilepsies. The emerging research developed in recent years has allowed us to identify the main molecular pathways involved in these processes. These molecular pathways could constitute future therapeutic targets for epilepsy. Different drugs current or in development have demonstrated their capacity to inhibit or modulate molecular pathways involved in the immunologic or neuroinflammatory mechanisms described in epilepsy. Some of them should be tested in the future as possible antiepileptic drugs.
Collapse
Affiliation(s)
- María José Aguilar-Castillo
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Biotechnology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
| | - Pablo Cabezudo-García
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Nicolas Lundahl Ciano-Petersen
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Guillermina García-Martin
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Marta Marín-Gracia
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Guillermo Estivill-Torrús
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
| | - Pedro Jesús Serrano-Castro
- Epilepsy Unit, Regional University Hospital of Málaga, 29010 Málaga, Spain; (M.J.A.-C.); (P.C.-G.); (N.L.C.-P.); (G.G.-M.); (M.M.-G.); (G.E.-T.)
- Andalusian Network for Clinical and Translational Research in Neurology (Neuro-RECA), 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA), 29010 Málaga, Spain
- Neurology Service, Regional University Hospital of Málaga, 29010 Málaga, Spain
- Department of Medicine, University of Málaga, 29071 Málaga, Spain
| |
Collapse
|
80
|
Immune Dysregulation in Autism Spectrum Disorder: What Do We Know about It? Int J Mol Sci 2022; 23:ijms23063033. [PMID: 35328471 PMCID: PMC8955336 DOI: 10.3390/ijms23063033] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex multifactorial neurodevelopmental disorders characterized by a wide and variable set of neuropsychiatric symptoms, including deficits in social communication, narrow and restricted interests, and repetitive behavior. The immune hypothesis is considered to be a major factor contributing to autism pathogenesis, as well as a way to explain the differences of the clinical phenotypes and comorbidities influencing disease course and severity. Evidence highlights a link between immune dysfunction and behavioral traits in autism from several types of evidence found in both cerebrospinal fluid and peripheral blood and their utility to identify autistic subgroups with specific immunophenotypes; underlying behavioral symptoms are also shown. This review summarizes current insights into immune dysfunction in ASD, with particular reference to the impact of immunological factors related to the maternal influence of autism development; comorbidities influencing autism disease course and severity; and others factors with particular relevance, including obesity. Finally, we described main elements of similarities between immunopathology overlapping neurodevelopmental and neurodegenerative disorders, taking as examples autism and Parkinson Disease, respectively.
Collapse
|
81
|
The role of glial cells in multiple sclerosis disease progression. Nat Rev Neurol 2022; 18:237-248. [PMID: 35190704 DOI: 10.1038/s41582-022-00624-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 12/13/2022]
Abstract
Despite the development of highly effective treatments for relapsing-remitting multiple sclerosis (MS), limited progress has been made in addressing primary progressive or secondary progressive MS, both of which lead to loss of oligodendrocytes and neurons and axons, and to irreversible accumulation of disability. Neuroinflammation is central to all forms of MS. The current effective therapies for relapsing-remitting MS target the peripheral immune system; these treatments, however, have repeatedly failed in progressive MS. Greater understanding of inflammation driven by CNS-resident cells - including astrocytes and microglia - is, therefore, required to identify novel potential therapeutic opportunities. Advances in imaging, biomarker analysis and genomics suggest that microglia and astrocytes have central roles in the progressive disease process. In this Review, we provide an overview of the involvement of astrocytes and microglia at major sites of pathology in progressive MS. We discuss current and future therapeutic approaches to directly target glial cells, either to inhibit pathogenic functions or to restore homeostatic functions lost during the course of the disease. We also discuss how bidirectional communication between astrocytes and microglia needs to be considered, as therapeutic targeting of one is likely to alter the functions of the other.
Collapse
|
82
|
Jang KB, You MJ, Yang B, Rim C, Kim HJ, Kwon MS. Persistent Acidic Environment Induces Impaired Phagocytosis via ERK in Microglia. Neurochem Res 2022; 47:1341-1353. [PMID: 35103911 DOI: 10.1007/s11064-022-03533-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/28/2022]
Abstract
Acidic environment evoked by stroke, traumatic brain injury, and Alzheimer's disease may change the functional properties of microglia. Nevertheless, the underlying mechanisms of functional changes in microglia remain unclear. In this study, we found that acidic stimuli (pH 6.8) increased rapidly interleukin (IL)-1β and IL-6 mRNA levels and subsequently reduced IL-10, transforming growth factor (TGF)-β1, Cx3cr1, and P2ry12 as the exposure time to acidic environment increase in BV2 cells. In addition, persistent acidic environment (pH 6.8 for 6 h) induced impaired phagocytic function in BV2 cells. Short-term acidic exposure (pH 6.8 for 30 min) increased cyclic AMP (cAMP) and phospho-protein kinase A (PKA) but inhibited phospho-extracellular signal-regulated kinase (p-ERK). However, under persistent acidic environment (pH 6.8 for 6 h), cyclic AMP and PKA were normalized and p-ERK was increased with TDAG8 (T cell death associated gene 8; GPR65) reduction. FR 180,204, an ERK inhibitor, rescued the persistent acidic environment-induced functional changes in BV2 cells and its effect was recapitulated in primary neonatal microglia. Thus, we propose that ERK targeting may be an alternative strategy to restore microglial dysfunction in the central nervous system (CNS) acidic environment in various neurological disorders.
Collapse
Affiliation(s)
- Kyu-Beom Jang
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Bohyun Yang
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Chan Rim
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Hui-Ju Kim
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA BIO COMPLEX, CHA University, 335 Pangyo, Bundang-gu, Gyeonggi-do, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
83
|
Silburt J, Aubert I. MORPHIOUS: an unsupervised machine learning workflow to detect the activation of microglia and astrocytes. J Neuroinflammation 2022; 19:24. [PMID: 35093113 PMCID: PMC8800241 DOI: 10.1186/s12974-021-02376-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background In conditions of brain injury and degeneration, defining microglial and astrocytic activation using cellular markers alone remains a challenging task. We developed the MORPHIOUS software package, an unsupervised machine learning workflow which can learn the morphologies of non-activated astrocytes and microglia, and from this information, infer clusters of microglial and astrocytic activation in brain tissue. Methods MORPHIOUS combines a one-class support vector machine with the density-based spatial clustering of applications with noise (DBSCAN) algorithm to identify clusters of microglial and astrocytic activation. Here, activation was triggered by permeabilizing the blood–brain barrier (BBB) in the mouse hippocampus using focused ultrasound (FUS). At 7 day post-treatment, MORPHIOUS was applied to evaluate microglial and astrocytic activation in histological tissue. MORPHIOUS was further evaluated on hippocampal sections of TgCRND8 mice, a model of amyloidosis that is prone to microglial and astrocytic activation. Results MORPHIOUS defined two classes of microglia, termed focal and proximal, that are spatially adjacent to the activating stimulus. Focal and proximal microglia demonstrated activity-associated features, including increased levels of ionized calcium-binding adapter molecule 1 expression, enlarged soma size, and deramification. MORPHIOUS further identified clusters of astrocytes characterized by activity-related changes in glial fibrillary acidic protein expression and branching. To validate these classifications following FUS, co-localization with activation markers were assessed. Focal and proximal microglia co-localized with the transforming growth factor beta 1, while proximal astrocytes co-localized with Nestin. In TgCRND8 mice, microglial and astrocytic activation clusters were found to correlate with amyloid-β plaque load. Thus, by only referencing control microglial and astrocytic morphologies, MORPHIOUS identified regions of interest corresponding to microglial and astrocytic activation. Conclusions Overall, our algorithm is a reliable and sensitive method for characterizing microglial and astrocytic activation following FUS-induced BBB permeability and in animal models of neurodegeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02376-9.
Collapse
|
84
|
Zhou K, Han J, Lund H, Boggavarapu NR, Lauschke VM, Goto S, Cheng H, Wang Y, Tachi A, Xie C, Zhu K, Sun Y, Osman AM, Liang D, Han W, Gemzell-Danielsson K, Betsholtz C, Zhang XM, Zhu C, Enge M, Joseph B, Harris RA, Blomgren K. An overlooked subset of Cx3cr1 wt/wt microglia in the Cx3cr1 CreER-Eyfp/wt mouse has a repopulation advantage over Cx3cr1 CreER-Eyfp/wt microglia following microglial depletion. J Neuroinflammation 2022; 19:20. [PMID: 35062962 PMCID: PMC8783445 DOI: 10.1186/s12974-022-02381-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/28/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Fluorescent reporter labeling and promoter-driven Cre-recombinant technologies have facilitated cellular investigations of physiological and pathological processes, including the widespread use of the Cx3cr1CreER-Eyfp/wt mouse strain for studies of microglia. METHODS Immunohistochemistry, Flow Cytometry, RNA sequencing and whole-genome sequencing were used to identify the subpopulation of microglia in Cx3cr1CreER-Eyfp/wt mouse brains. Genetically mediated microglia depletion using Cx3cr1CreER-Eyfp/wtRosa26DTA/wt mice and CSF1 receptor inhibitor PLX3397 were used to deplete microglia. Primary microglia proliferation and migration assay were used for in vitro studies. RESULTS We unexpectedly identified a subpopulation of microglia devoid of genetic modification, exhibiting higher Cx3cr1 and CX3CR1 expression than Cx3cr1CreER-Eyfp/wtCre+Eyfp+ microglia in Cx3cr1CreER-Eyfp/wt mouse brains, thus termed Cx3cr1highCre-Eyfp- microglia. This subpopulation constituted less than 1% of all microglia under homeostatic conditions, but after Cre-driven DTA-mediated microglial depletion, Cx3cr1highCre-Eyfp- microglia escaped depletion and proliferated extensively, eventually occupying one-third of the total microglial pool. We further demonstrated that the Cx3cr1highCre-Eyfp- microglia had lost their genetic heterozygosity and become homozygous for wild-type Cx3cr1. Therefore, Cx3cr1highCre-Eyfp- microglia are Cx3cr1wt/wtCre-Eyfp-. Finally, we demonstrated that CX3CL1-CX3CR1 signaling regulates microglial repopulation both in vivo and in vitro. CONCLUSIONS Our results raise a cautionary note regarding the use of Cx3cr1CreER-Eyfp/wt mouse strains, particularly when interpreting the results of fate mapping, and microglial depletion and repopulation studies.
Collapse
Affiliation(s)
- Kai Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China.
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| | - Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Neuroimmunology Center, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Harald Lund
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Shinobu Goto
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Nagoya City University Hospital, Nagoya, Japan
| | - Huaitao Cheng
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yuyu Wang
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Asuka Tachi
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Cuicui Xie
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ahmed M Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Dong Liang
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Wei Han
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Gemzell-Danielsson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO-Centre, Karolinska University Hospital, Stockholm, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, Campus Flemingsberg, Huddinge, Sweden
| | - Xing-Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Changlian Zhu
- Centre for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, The Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Martin Enge
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Bertrand Joseph
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, The Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou University, Zhengzhou, China.
- Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
85
|
Yang B, Ryu JS, Rim C, Shin JU, Kwon MS. Possible role of arginase 1 positive microglia on depressive/anxiety-like behaviors in atopic dermatitis mouse model. Arch Pharm Res 2022; 45:11-28. [DOI: 10.1007/s12272-022-01369-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
|
86
|
Çalışkan G, French T, Enrile Lacalle S, Del Angel M, Steffen J, Heimesaat MM, Rita Dunay I, Stork O. Antibiotic-induced gut dysbiosis leads to activation of microglia and impairment of cholinergic gamma oscillations in the hippocampus. Brain Behav Immun 2022; 99:203-217. [PMID: 34673174 DOI: 10.1016/j.bbi.2021.10.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022] Open
Abstract
Antibiotics are widely applied for the treatment of bacterial infections, but their long-term use may lead to gut flora dysbiosis and detrimental effects on brain physiology, behavior as well as cognitive performance. Still, a striking lack of knowledge exists concerning electrophysiological correlates of antibiotic-induced changes in gut microbiota and behavior. Here, we investigated changes in the synaptic transmission and plasticity together with behaviorally-relevant network activities from the hippocampus of antibiotic-treated mice. Prolonged antibiotic treatment led to a reduction of myeloid cell pools in bone marrow, circulation and those surveilling the brain. Circulating Ly6Chi inflammatory monocytes adopted a proinflammatory phenotype with increased expression of CD40 and MHC II. In the central nervous system, microglia displayed a subtle activated phenotype with elevated CD40 and MHC II expression, increased IL-6 and TNF production as well as with an increased number of Iba1 + cells in the hippocampal CA3 and CA1 subregions. Concomitantly, we detected a substantial reduction in the synaptic transmission in the hippocampal CA1 after antibiotic treatment. In line, carbachol-induced cholinergic gamma oscillation were reduced upon antibiotic treatment while the incidence of hippocampal sharp waves was elevated. These alterations were associated with the global changes in the expression of neurotrophin nerve growth factor and inducible nitric oxide synthase, both of which have been shown to influence cholinergic system in the hippocampus. Overall, our study demonstrates that antibiotic-induced dysbiosis of the gut microbiome and subsequent alteration of the immune cell function are associated with reduced synaptic transmission and gamma oscillations in the hippocampus, a brain region that is critically involved in mediation of innate and cognitive behavior.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| | - Timothy French
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| | | | - Miguel Del Angel
- Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Ildiko Rita Dunay
- Center for Behavioral Brain Sciences, Magdeburg, Germany; Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| | - Oliver Stork
- Institute of Biology, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
87
|
Logiacco F, Xia P, Georgiev SV, Franconi C, Chang YJ, Ugursu B, Sporbert A, Kühn R, Kettenmann H, Semtner M. Microglia sense neuronal activity via GABA in the early postnatal hippocampus. Cell Rep 2021; 37:110128. [PMID: 34965412 DOI: 10.1016/j.celrep.2021.110128] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/14/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023] Open
Abstract
Microglia, the resident macrophages in the central nervous system, express receptors for classical neurotransmitters, such as γ-aminobutyric acid (GABA) and glutamate, suggesting that they sense synaptic activity. To detect microglial Ca2+ responses to neuronal activity, we generate transgenic mouse lines expressing the fluorescent Ca2+ indicator GCaMP6m, specifically in microglia and demonstrate that electrical stimulation of the Schaffer collateral pathway results in microglial Ca2+ responses in early postnatal but not adult hippocampus. Preceding the microglial responses, we also observe similar Ca2+ responses in astrocytes, and both are sensitive to tetrodotoxin. Blocking astrocytic glutamate uptake or GABA transport abolishes stimulation-induced microglial responses as well as antagonizing the microglial GABAB receptor. Our data, therefore, suggest that the neuronal activity-induced glutamate uptake and the release of GABA by astrocytes trigger the activation of GABAB receptors in microglia. This neuron, astrocyte, and microglia communication pathway might modulate microglial activity in developing neuronal networks.
Collapse
Affiliation(s)
- Francesca Logiacco
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 12169 Berlin, Germany
| | - Pengfei Xia
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Svilen Veselinov Georgiev
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Celeste Franconi
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Yi-Jen Chang
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Bilge Ugursu
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Ralf Kühn
- Transgenic Core Facility, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| |
Collapse
|
88
|
Hamner MA, McDonough A, Gong DC, Todd LJ, Rojas G, Hodecker S, Ransom CB, Reh TA, Ransom BR, Weinstein JR. Microglial depletion abolishes ischemic preconditioning in white matter. Glia 2021; 70:661-674. [PMID: 34939240 DOI: 10.1002/glia.24132] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 01/17/2023]
Abstract
Ischemic preconditioning (IPC) is a phenomenon whereby a brief, non-injurious ischemic exposure enhances tolerance to a subsequent ischemic challenge. The mechanism of IPC has mainly been studied in rodent stroke models where gray matter (GM) constitutes about 85% of the cerebrum. In humans, white matter (WM) is 50% of cerebral volume and is a critical component of stroke damage. We developed a novel CNS WM IPC model using the mouse optic nerve (MON) and identified the involved immune signaling pathways. Here we tested the hypothesis that microglia are necessary for WM IPC. Microglia were depleted by treatment with the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX5622. MONs were exposed to transient ischemia in vivo, acutely isolated 72 h later, and subjected to oxygen-glucose deprivation (OGD) to simulate a severe ischemic injury (i.e., stroke). Functional and structural axonal recovery was assessed by recording compound action potentials (CAPs) and by microscopy using quantitative stereology. Microglia depletion eliminated IPC-mediated protection. In control mice, CAP recovery was improved in preconditioned MONs compared with non-preconditioned MONs, however, in PLX5622-treated mice, we observed no difference in CAP recovery between preconditioned and non-preconditioned MONs. Microgliadepletion also abolished IPC protective effects on axonal integrity and survival of mature (APC+ ) oligodendrocytes after OGD. IPC-mediated protection was independent of retinal injury suggesting it results from mechanistic processes intrinsic to ischemia-exposed WM. We conclude that preconditioned microglia are critical for IPC in WM. The "preconditioned microglia" phenotype might protect against other CNS pathologies and is a neurotherapeutic horizon worth exploring.
Collapse
Affiliation(s)
- Margaret A Hamner
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Ashley McDonough
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Davin C Gong
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Levi J Todd
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, Washington, USA
| | - German Rojas
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Sibylle Hodecker
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Christopher B Ransom
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Thomas A Reh
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Bruce R Ransom
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA.,Neuroscience Department, City University of Hong Kong, Kowloon, Hong Kong
| | - Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA.,Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
89
|
Arrieta VA, Najem H, Petrosyan E, Lee-Chang C, Chen P, Sonabend AM, Heimberger AB. The Eclectic Nature of Glioma-Infiltrating Macrophages and Microglia. Int J Mol Sci 2021; 22:13382. [PMID: 34948178 PMCID: PMC8705822 DOI: 10.3390/ijms222413382] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/23/2022] Open
Abstract
Glioblastomas (GBMs) are complex ecosystems composed of highly multifaceted tumor and myeloid cells capable of responding to different environmental pressures, including therapies. Recent studies have uncovered the diverse phenotypical identities of brain-populating myeloid cells. Differences in the immune proportions and phenotypes within tumors seem to be dictated by molecular features of glioma cells. Furthermore, increasing evidence underscores the significance of interactions between myeloid cells and glioma cells that allow them to evolve in a synergistic fashion to sustain tumor growth. In this review, we revisit the current understanding of glioma-infiltrating myeloid cells and their dialogue with tumor cells in consideration of their increasing recognition in response and resistance to immunotherapies as well as the immune impact of the current chemoradiotherapy used to treat gliomas.
Collapse
Affiliation(s)
- Víctor A. Arrieta
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico
| | - Hinda Najem
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
| | - Edgar Petrosyan
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
| | - Catalina Lee-Chang
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
| | - Peiwen Chen
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
| | - Adam M. Sonabend
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
| | - Amy B. Heimberger
- Department of Neurosurgery, Lou and Jean Malnati Brain Tumor Institute, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (V.A.A.); (H.N.); (E.P.); (C.L.-C.); (P.C.)
| |
Collapse
|
90
|
Crosstalk between Neuron and Glial Cells in Oxidative Injury and Neuroprotection. Int J Mol Sci 2021; 22:ijms222413315. [PMID: 34948108 PMCID: PMC8709409 DOI: 10.3390/ijms222413315] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/30/2022] Open
Abstract
To counteract oxidative stress and associated brain diseases, antioxidant systems rescue neuronal cells from oxidative stress by neutralizing reactive oxygen species and preserving gene regulation. It is necessary to understand the communication and interactions between brain cells, including neurons, astrocytes and microglia, to understand oxidative stress and antioxidant mechanisms. Here, the role of glia in the protection of neurons against oxidative injury and glia–neuron crosstalk to maintain antioxidant defense mechanisms and brain protection are reviewed. The first part of this review focuses on the role of glia in the morphological and physiological changes required for brain homeostasis under oxidative stress and antioxidant defense mechanisms. The second part focuses on the essential crosstalk between neurons and glia for redox balance in the brain for protection against oxidative stress.
Collapse
|
91
|
van der Wildt B, Janssen B, Pekošak A, Stéen EJL, Schuit RC, Kooijman EJM, Beaino W, Vugts DJ, Windhorst AD. Novel Thienopyrimidine-Based PET Tracers for P2Y 12 Receptor Imaging in the Brain. ACS Chem Neurosci 2021; 12:4465-4474. [PMID: 34757711 PMCID: PMC8640995 DOI: 10.1021/acschemneuro.1c00641] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
The P2Y12 receptor (P2Y12R) is uniquely expressed
on microglia in the brain, and its expression level directly depends
on the microglial activation state. Therefore, P2Y12R provides
a promising imaging marker for distinguishing the pro- and anti-inflammatory
microglial phenotypes, both of which play crucial roles in neuroinflammatory
diseases. In this study, three P2Y12R antagonists were
selected from the literature, radiolabeled with carbon-11 or fluorine-18,
and evaluated in healthy Wistar rats. Brain imaging was performed
with and without blocking of efflux transporters P-glycoprotein and breast cancer resistance protein using tariquidar.
Low brain uptake in healthy rats was observed for all tracers at baseline
conditions, whereas blocking of efflux transporters resulted in a
strong (6–7 fold) increase in brain uptake for both of them.
Binding of the most promising tracer, [18F]3, was further evaluated by in vitro autoradiography on rat brain
sections, ex vivo metabolite studies, and in vivo P2Y12R blocking studies. In vitro binding of [18F]3 on rat brain sections indicated high P2Y12R targeting
with approximately 70% selective and specific binding. At 60 min post-injection,
over 95% of radioactivity in the brain accounted for an intact tracer.
In blood plasma, still 40% intact tracer was found, and formed metabolites
did not enter the brain. A moderate P2Y12R blocking effect
was observed in vivo by positron emission tomography (PET) imaging
with [18F]3 (p = 0.04). To
conclude, three potential P2Y12R PET tracers were obtained
and analyzed for P2Y12R targeting in the brain. Unfortunately,
the brain uptake appeared low. Future work will focus on the design
of P2Y12R inhibitors with improved physicochemical characteristics
to reduce efflux transport and increase brain penetration.
Collapse
Affiliation(s)
- Berend van der Wildt
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Bieneke Janssen
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Aleksandra Pekošak
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - E. Johanna L. Stéen
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Robert C. Schuit
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Esther J. M. Kooijman
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Wissam Beaino
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Danielle J. Vugts
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Albert D. Windhorst
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
92
|
New Insights into the Role of Cysteine Cathepsins in Neuroinflammation. Biomolecules 2021; 11:biom11121796. [PMID: 34944440 PMCID: PMC8698589 DOI: 10.3390/biom11121796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, which is mediated by microglia and astrocytes, is associated with the progression of neurodegenerative diseases. Increasing evidence shows that activated microglia induce the expression and secretion of various lysosomal cathepsins, particularly during the early stage of neuroinflammation. This trigger signaling cascade that aggravate neurodegeneration. To date, most research on neuroinflammation has focused on the role of cysteine cathepsins, the largest cathepsin family. Cysteine cathepsins are primarily responsible for protein degradation in lysosomes; however, they also play a role in regulating a number of other important physiological and pathological processes. This review focuses on the functional roles of cysteine cathepsins in the central nervous system during neuroinflammation, with an emphasis on their roles in the polarization of microglia and neuroinflammation signaling, which in turn causes neuronal death and thus neurodegeneration.
Collapse
|
93
|
Wang YJ, Downey MA, Choi S, Shoup TM, Elmaleh DR. Cromolyn platform suppresses fibrosis and inflammation, promotes microglial phagocytosis and neurite outgrowth. Sci Rep 2021; 11:22161. [PMID: 34772945 PMCID: PMC8589953 DOI: 10.1038/s41598-021-00465-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuroinflammation and may perpetuate ongoing fibrotic reactions within the central nervous system. Unfortunately, there is no therapeutic available that treats neurodegenerative inflammation and its sequelae. Here we utilize cromolyn, a mast cell inhibitor with anti-inflammatory capabilities, and its fluorinated analogue F-cromolyn to study fibrosis-related protein regulation and secretion downstream of neuroinflammation and their ability to promote microglial phagocytosis and neurite outgrowth. In this report, RNA-seq analysis shows that administration of the pro-inflammatory cytokine TNF-α to HMC3 human microglia results in a robust upregulation of fibrosis-associated genes. Subsequent treatment with cromolyn and F-cromolyn resulted in reduced secretion of collagen XVIII, fibronectin, and tenascin-c. Additionally, we show that cromolyn and F-cromolyn reduce pro-inflammatory proteins PLP1, PELP1, HSP90, IL-2, GRO-α, Eotaxin, and VEGF-Α, while promoting secretion of anti-inflammatory IL-4 in HMC3 microglia. Furthermore, cromolyn and F-cromolyn augment neurite outgrowth in PC12 neuronal cells in concert with nerve growth factor. Treatment also differentially altered secretion of neurogenesis-related proteins TTL, PROX1, Rab35, and CSDE1 in HMC3 microglia. Finally, iPSC-derived human microglia more readily phagocytose Aβ42 with cromolyn and F-cromolyn relative to controls. We propose the cromolyn platform targets multiple proteins upstream of PI3K/Akt/mTOR, NF-κB, and GSK-3β signaling pathways to affect cytokine, chemokine, and fibrosis-related protein expression.
Collapse
Affiliation(s)
| | | | - Sungwoon Choi
- Department of New Drug Discovery, Chungnam National University, Daejeon, South Korea
| | - Timothy M Shoup
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA
| | - David R Elmaleh
- AZTherapies, Inc., Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA.
| |
Collapse
|
94
|
Song Y, Wu Z, Zhao P. The protective effects of activating Sirt1/NF-κB pathway for neurological disorders. Rev Neurosci 2021; 33:427-438. [PMID: 34757706 DOI: 10.1515/revneuro-2021-0118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/08/2021] [Indexed: 12/30/2022]
Abstract
Sirt1, a member of the sirtuins family, is a nicotinamide adenosine dinucleotide (NAD+)-dependent deacetylase. It can be involved in the regulation of several processes including inflammatory response, apoptosis, oxidative stress, energy metabolism, and autophagy by exerting deacetylation. Nuclear factor-κB (NF-κB), a crucial nuclear transcription factor with specific DNA binding sequences, exists in almost all cells and plays a vital role in several biological processes involving inflammatory response, immune response, and apoptosis. As the hub of multiple intracellular signaling pathways, the activity of NF-κB is regulated by multiple factors. Sirt1 can both directly deacetylate NF-κB and indirectly through other molecules to inhibit its activity. We would like to emphasize that Sirt1/NF-κB is a signaling pathway that is closely related to neuroinflammation. Many recent studies have demonstrated the neuroprotective effects of Sirt1/NF-κB signaling pathway activation applied to the treatment of neurological related diseases. In this review, we focus on new advances in the neuroprotective effects of the Sirt1/NF-κB pathway. First, we briefly review Sirt1 and NF-κB, two key molecules of cellular metabolism. Next, we discuss the connection between NF-κB and neuroinflammation. In addition, we explore how Sirt1 regulates NF-κB in nerve cells and relevant evidence. Finally, we analyze the therapeutic effects of the Sirt1/NF-κB pathway in several common neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
95
|
Olajide OA, Iwuanyanwu VU, Adegbola OD, Al-Hindawi AA. SARS-CoV-2 Spike Glycoprotein S1 Induces Neuroinflammation in BV-2 Microglia. Mol Neurobiol 2021; 59:445-458. [PMID: 34709564 PMCID: PMC8551352 DOI: 10.1007/s12035-021-02593-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023]
Abstract
In addition to respiratory complications produced by SARS‐CoV‐2, accumulating evidence suggests that some neurological symptoms are associated with the disease caused by this coronavirus. In this study, we investigated the effects of the SARS‐CoV‐2 spike protein S1 stimulation on neuroinflammation in BV-2 microglia. Analyses of culture supernatants revealed an increase in the production of TNF-α, IL-6, IL-1β and iNOS/NO. S1 also increased protein levels of phospho-p65 and phospho-IκBα, as well as enhanced DNA binding and transcriptional activity of NF-κB. These effects of the protein were blocked in the presence of BAY11-7082 (1 µM). Exposure of S1 to BV-2 microglia also increased the protein levels of NLRP3 inflammasome and enhanced caspase-1 activity. Increased protein levels of p38 MAPK was observed in BV-2 microglia stimulated with the spike protein S1 (100 ng/ml), an action that was reduced in the presence of SKF 86,002 (1 µM). Results of immunofluorescence microscopy showed an increase in TLR4 protein expression in S1-stimulated BV-2 microglia. Furthermore, pharmacological inhibition with TAK 242 (1 µM) and transfection with TLR4 small interfering RNA resulted in significant reduction in TNF-α and IL-6 production in S1-stimulated BV-2 microglia. These results have provided the first evidence demonstrating S1-induced neuroinflammation in BV-2 microglia. We propose that induction of neuroinflammation by this protein in the microglia is mediated through activation of NF-κB and p38 MAPK, possibly as a result of TLR4 activation. These results contribute to our understanding of some of the mechanisms involved in CNS pathologies of SARS-CoV-2.
Collapse
Affiliation(s)
- Olumayokun A Olajide
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK.
| | - Victoria U Iwuanyanwu
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Oyinkansola D Adegbola
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Alaa A Al-Hindawi
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| |
Collapse
|
96
|
Hambali A, Kumar J, Hashim NFM, Maniam S, Mehat MZ, Cheema MS, Mustapha M, Adenan MI, Stanslas J, Hamid HA. Hypoxia-Induced Neuroinflammation in Alzheimer's Disease: Potential Neuroprotective Effects of Centella asiatica. Front Physiol 2021; 12:712317. [PMID: 34721056 PMCID: PMC8551388 DOI: 10.3389/fphys.2021.712317] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterised by the presence of extracellular beta-amyloid fibrillary plaques and intraneuronal neurofibrillary tau tangles in the brain. Recurring failures of drug candidates targeting these pathways have prompted research in AD multifactorial pathogenesis, including the role of neuroinflammation. Triggered by various factors, such as hypoxia, neuroinflammation is strongly linked to AD susceptibility and/or progression to dementia. Chronic hypoxia induces neuroinflammation by activating microglia, the resident immune cells in the brain, along with an increased in reactive oxygen species and pro-inflammatory cytokines, features that are common to many degenerative central nervous system (CNS) disorders. Hence, interests are emerging on therapeutic agents and plant derivatives for AD that target the hypoxia-neuroinflammation pathway. Centella asiatica is one of the natural products reported to show neuroprotective effects in various models of CNS diseases. Here, we review the complex hypoxia-induced neuroinflammation in the pathogenesis of AD and the potential application of Centella asiatica as a therapeutic agent in AD or dementia.
Collapse
Affiliation(s)
- Aqilah Hambali
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sandra Maniam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Manraj Singh Cheema
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Johnson Stanslas
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
97
|
Aubert A, Stüder F, Colombo BM, Mendoza-Parra MA. A Core Transcription Regulatory Circuitry Defining Microglia Cell Identity Inferred from the Reanalysis of Multiple Human Microglia Differentiation Protocols. Brain Sci 2021; 11:brainsci11101338. [PMID: 34679401 PMCID: PMC8533937 DOI: 10.3390/brainsci11101338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022] Open
Abstract
Microglia, the immune cells in the brain involved in both homeostasis and injury/infection control, play a predominant role in neurodegenerative diseases. In vivo studies on microglia are limited due to the requirement of surgical intervention, which can lead to the destruction of the tissues. Over the last few years, multiple protocols-presenting a variety of strategies-have described microglia differentiation issued from human pluripotent stem cells. Herein, we have reanalyzed the transcriptomes released on six different microglia differentiation protocols and revealed a consensus core of master transcription regulatory circuitry defining microglia identity. Furthermore, we have discussed the major divergencies among the studied protocols and have provided suggestions to further enhance microglia differentiation assays.
Collapse
|
98
|
Hineno A, Oyanagi K, Yoshida T, Sakai Y, Kanno H, Sekijima Y. Spread of vimentin-immunoreactive cells within the plaque-like lesion in the spinal anterior horn of a patient with post-poliomyelitis syndrome. Neuropathology 2021; 41:406-411. [PMID: 34541709 DOI: 10.1111/neup.12768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 11/27/2022]
Abstract
A Japanese man in the present study experienced acute weakness in his right leg as a two year old. The strength in his leg gradually recovered and developed, and he could play golf and climb mountains up to around the age of 50. From approximately 55 years of age, he became unable to stand up from a stooped position. Muscle weakness and atrophy spread to his right arm, and an electromyography revealed a neurogenic pattern in his lower and upper extremities. The patient was diagnosed as having post-poliomyelitis syndrome (PPS). Numbness in both the legs and pain in the buttocks occurred after 60 years of age. Computed tomography and magnetic resonance imaging at that time revealed spondylosis and protrusion of an osteophye in lower thoracic vertebrae compressing the second lumbar segment of the spinal cord. He died of malignant lymphoma and acute interstitial pneumonia at 80 years of age. Pathological examination revealed transverse myelopathy at the second lumbar segment of the spinal cord and total necrosis. The anterior horn and the intermediate zone of the third and fourth lumbar segments of the spinal cord on the right side were atrophic and diffusely gliotic. An oval-shaped plaque-like lesion was observed in the right anterior horn at the third and fourth lumbar segments of the spinal cord. Neurons and synaptophysin immunoreactivity had completely disappeared in the plaque-like lesion. A striking spread of vimentin-immunoreactive cells was found corresponding to the lesion, while glial fibrillary acidic protein-immunoreactive astrocytes existed evenly in the anterior horn and intermediate zone on both sides of the third and fourth lumber segments of the spinal cord. Virological examination using the autopsied materials was negative for poliovirus. Neither transactivation response DNA-binding protein of 43 kDa-immunoreactive inclusion nor Bunina body was seen in the spinal cord. The present paper demonstrates new findings of a noteworthy response of the vimentin-immunoreactive cells within the peculiar "plaque-like lesion" in the PPS.
Collapse
Affiliation(s)
- Akiyo Hineno
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan.,Intractable Disease Care Center, Shinshu University Hospital, Matsumoto, Japan
| | - Kiyomitsu Oyanagi
- Department of Brain Disease Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takuhiro Yoshida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Yasuhiro Sakai
- Department of Molecular Pathology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Hiroyuki Kanno
- Department of Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
99
|
Sahu R, Upadhayay S, Mehan S. Inhibition of extracellular regulated kinase (ERK)-1/2 signaling pathway in the prevention of ALS: Target inhibitors and influences on neurological dysfunctions. Eur J Cell Biol 2021; 100:151179. [PMID: 34560374 DOI: 10.1016/j.ejcb.2021.151179] [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: 07/28/2021] [Revised: 08/18/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cell signal transduction pathways are essential modulators of several physiological and pathological processes in the brain. During overactivation, these signaling processes may lead to disease progression. Abnormal protein kinase activation is associated with several biological dysfunctions that facilitate neurodegeneration under different biological conditions. As a result, these signaling pathways are essential in understanding brain disorders' development or progression. Recent research findings indicate the crucial role of extracellular signal-regulated kinase-1/2 (ERK-1/2) signaling during the neuronal development process. ERK-1/2 is a key component of its mitogen-activated protein kinase (MAPK) group, controlling certain neurological activities by regulating metabolic pathways, cell proliferation, differentiation, and apoptosis. ERK-1/2 also influences neuronal elastic properties, nerve growth, and neurological and cognitive processing during brain injuries. The primary goal of this review is to elucidate the activation of ERK1/2 signaling, which is involved in the development of several ALS-related neuropathological dysfunctions. ALS is a rare neurological disorder category that mainly affects the nerve cells responsible for regulating voluntary muscle activity. ALS is progressive, which means that the symptoms are getting worse over time, and there is no cure for ALS and no effective treatment to avoid or reverse. Genetic abnormalities, oligodendrocyte degradation, glial overactivation, and immune deregulation are associated with ALS progression. Furthermore, the current review also identifies ERK-1/2 signaling inhibitors that can promote neuroprotection and neurotrophic effects against the clinical-pathological presentation of ALS. As a result, in the future, the potential ERK-1/2 signaling inhibitors could be used in the treatment of ALS and related neurocomplications.
Collapse
Affiliation(s)
- Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shubham Upadhayay
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
100
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|