1
|
Tripathi A, Rai NK, Perles A, Courtney H, Jones C, Sapra A, Plemel J, Dutta R. Dicer deficiency affects microglial function during demyelination and impairs remyelination. Neurobiol Dis 2025; 208:106879. [PMID: 40120829 PMCID: PMC12068937 DOI: 10.1016/j.nbd.2025.106879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Microglia are essential regulators of central nervous system (CNS) homeostasis, playing key roles in demyelination and remyelination. Dysregulated microglial activity contributes to pathological inflammation and impaired repair processes in demyelinating diseases. Here, we investigate the role of Dicer1, a critical enzyme in microRNA biogenesis, in affecting microglial function, demyelination, and remyelination. Loss of Dicer1 in microglia resulted in amplified inflammatory responses, defective myelin debris clearance, and disruption of metabolic homeostasis, leading to exacerbated demyelination and delayed remyelination. Transcriptomic analysis revealed significant upregulation of inflammatory pathways, including interferon signaling and JAK/STAT activation, alongside a loss of homeostatic microglial gene expression. Protein-level validation confirmed sustained secretion of pro-inflammatory cytokines such as IFN-γ, IL-16, and CXCL12, creating a chronic inflammatory environment that impaired remyelination. Furthermore, Dicer1-deficient microglia failed to support oligodendrocyte progenitor cells (OPCs) differentiation/maturation, with increased apoptosis of mature oligodendrocytes (OLs), contributing to remyelination failure. These findings identify Dicer1 as a critical regulator of microglial homeostasis and inflammation resolution, highlighting its potential as a therapeutic target to mitigate inflammation and promote repair in demyelinating diseases.
Collapse
Affiliation(s)
- Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | | | - Aaron Perles
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Haley Courtney
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Claire Jones
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Adya Sapra
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Jason Plemel
- Neuroscience and Mental Health Institute, Department of Medicine, Division of Neurology, Department of Medical Microbiology and Immunology, University of Alberta, Canada
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA; Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.
| |
Collapse
|
2
|
Wang Z, Li W, Li J, Jin T, Chen H, Liang F, Liu S, Jia J, Liu T, Liu Y, Yu L, Xue X, Zhao J, Huang T, Huang X, Wang H, Li Y, Luo B, Zhang Z. Neutrophil-modulated Dicer expression in macrophages influences inflammation resolution. Cell Mol Life Sci 2025; 82:114. [PMID: 40074991 PMCID: PMC11904050 DOI: 10.1007/s00018-025-05644-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/09/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
The precise molecular mechanisms through which neutrophils regulate macrophages in the progression and resolution of acute inflammation remain poorly understood. Here, we present new findings on the role of Dicer in regulating macrophage phenotypic transitions essential for proper inflammatory progression and resolution, influenced by neutrophils. Using a zymosan A (Zym A)-induced self-limited mouse peritonitis model, we observed that Dicer expression in macrophages was significantly reduced by neutrophil-derived IFN-γ during the progression phase, but gradually returned to normal levels during the resolution phase following the engulfment of apoptotic neutrophils. Our study on macrophage-specific Dicer1-depletion (Dicer1-CKO) mice demonstrated that inflammation in these mice was more severe during the progression phase, characterized by increased pro-inflammatory cytokines and enhanced neutrophil trafficking. Additionally, resolution was impaired in Dicer1-CKO mice, leading to the accumulation of uncleared apoptotic neutrophils. Specifically, the absence of Dicer in macrophages resulted in M1 polarization and heightened bactericidal activity, facilitating the progression of acute inflammation. Conversely, inducing Dicer expression promoted macrophage transition to M2 polarization, enhancing apoptotic cell clearance and expediting the resolution of inflammation. Our findings suggest that Dicer plays a central role in regulating the progression and resolution of acute inflammation, with implications for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Zhishang Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Wenhua Li
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jia Li
- Department of Nephrology, Navy 971 Hospital, 22 Minjiang Road, Qingdao, Shandong, 266000, China
| | - Tianrong Jin
- Medical College of Chongqing University, Chongqing, 400030, China
| | - Hong Chen
- Department of Orthopedics, No. 903 Hospital of PLA Joint Logistic Support Force, 14 Lingyin Road, Lingyin Street, Xihu District, Hangzhou, Zhejiang, 310000, China
| | - Feihong Liang
- Department of Medical Science, Shunde Polytechnic, Foshan, China
| | - Shengran Liu
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Jialin Jia
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Tingting Liu
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Yu Liu
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China
| | - Liming Yu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Xiaodong Xue
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Jikai Zhao
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Tao Huang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Xinyi Huang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Huishan Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China.
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Bangwei Luo
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China.
| | - Zhiren Zhang
- Institute of Immunology, Army Medical University, 30 Gaotanyan Main Street, Chongqing, 400038, China.
| |
Collapse
|
3
|
Duarte T, Rassi DM, Carvalho A, Santos DE, Zanon S, Lucas G. Dysregulation of MicroRNA Biogenesis Machinery in Nervous System Diseases. Eur J Neurosci 2025; 61:e70058. [PMID: 40082738 DOI: 10.1111/ejn.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
MicroRNAs (miRNAs) have become essential modulators in many brain disorders, such as neurodegenerative diseases, psychiatry disorders, and chronic pain syndromes, and they play a critical role in controlling gene expression. This review investigates how disorders of the nervous system and pain research are affected by malfunctions in the miRNA biogenesis machinery. Despite tremendous progress, we still do not fully understand how these molecular regulators affect neuropathological processes. Even with the increasing amount of research, little is known about the malfunctions of the miRNA machinery, especially when it comes to the nervous system and the diseases that are linked to it. The results of recent research are compiled in this review, which emphasizes the role that disruptions in miRNA processing enzymes, including Drosha, Dicer, Argonaute, and RISC proteins, play in neurological conditions like Parkinson's and Alzheimer's diseases, as well as more general neurodegeneration. We also go over current studies on the stimulus-dependent, temporal, and spatial expression patterns of these essential miRNA biogenesis components in pain. These discoveries broaden our knowledge of the fundamental processes behind pain-related illnesses and present prospective directions for focused therapeutic approaches.
Collapse
Affiliation(s)
- Terence Duarte
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Diane Meyre Rassi
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Andrea Carvalho
- Department of Experimental Psychology, Neuroscience and Behavior Training Program, Institute of Psychology, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Sonia Zanon
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Guilherme Lucas
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
- Department of Experimental Psychology, Neuroscience and Behavior Training Program, Institute of Psychology, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
4
|
Raak SB, Hanley JG, O'Donnell C. Competition effects regulating the composition of the microRNA pool. J R Soc Interface 2025; 22:20240870. [PMID: 39965642 PMCID: PMC11835486 DOI: 10.1098/rsif.2024.0870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
MicroRNAS (miRNAs) are short non-coding RNAs that can repress mRNA translation to regulate protein synthesis. During their maturation, multiple types of pre-miRNAs compete for a shared pool of the enzyme Dicer. It is unknown how this competition for a shared resource influences the relative expression of mature miRNAs. We study this process in a computational model of pre-miRNA maturation, fitted to in vitro Drosophila S2 cell data. We find that those pre-miRNAs that efficiently interact with Dicer outcompete other pre-miRNAs, when Dicer is scarce. To test our model predictions, we re-analysed previously published ex vivo mouse striatum data with reduced Dicer1 expression. We calculated a proxy measure for pre-miRNA affinity to TRBP (a protein that loads pre-miRNAs to Dicer). This measures well-predicted mature miRNA levels in the data, validating our assumptions. We used this as a basis to test the the model's predictions through further analysis of the data. We found that pre-miRNAs with strong TRBP association are over-represented in competition conditions, consistent with the modelling. Finally using further simulations, we discovered that pre-miRNAs with low maturation rates can affect the mature miRNA pool via competition among pre-miRNAs. Overall, this work presents evidence of pre-miRNA competition regulating the composition of mature miRNAs.
Collapse
Affiliation(s)
- Sofia B. Raak
- School of Biochemistry, University of Bristol, University Walk, Clifton, BristolBS8 1TD, UK
| | - Jonathan G. Hanley
- School of Biochemistry, University of Bristol, University Walk, Clifton, BristolBS8 1TD, UK
| | - Cian O'Donnell
- School of Engineering Mathematics and Technology, University of Bristol, University Walk, Clifton, BristolBS8 1TD, UK
- School of Computing, Engineering and Intelligent Systems, Ulster University, Derry/LondonderryBT48 7JL, UK
| |
Collapse
|
5
|
Li Z, Mao K, Liu L, Xu S, Zeng M, Fu Y, Huang J, Li T, Gao G, Teng ZQ, Sun Q, Chen D, Cheng Y. Nuclear microRNA-mediated transcriptional control determines adult microglial homeostasis and brain function. Cell Rep 2024; 43:113964. [PMID: 38489263 DOI: 10.1016/j.celrep.2024.113964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/01/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
Microglia are versatile regulators in brain development and disorders. Emerging evidence links microRNA (miRNA)-mediated regulation to microglial function; however, the exact underlying mechanism remains largely unknown. Here, we uncover the enrichment of miR-137, a neuropsychiatric-disorder-associated miRNA, in the microglial nucleus, and reveal its unexpected nuclear functions in maintaining the microglial global transcriptomic state, phagocytosis, and inflammatory response. Mechanistically, microglial Mir137 deletion increases chromatin accessibility, which contains binding motifs for the microglial master transcription factor Pu.1. Through biochemical and bioinformatics analyses, we propose that miR-137 modulates Pu.1-mediated gene expression by suppressing Pu.1 binding to chromatin. Importantly, we find that increased Pu.1 binding upregulates the target gene Jdp2 (Jun dimerization protein 2) and that knockdown of Jdp2 significantly suppresses the impaired phagocytosis and pro-inflammatory response in Mir137 knockout microglia. Collectively, our study provides evidence supporting the notion that nuclear miR-137 acts as a transcriptional modulator and that this microglia-specific function is essential for maintaining normal adult brain function.
Collapse
Affiliation(s)
- Zhu Li
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Kexin Mao
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China; Southwest United Graduate School, Kunming 650500, China
| | - Lin Liu
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Shengyun Xu
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Min Zeng
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Yu Fu
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Jintao Huang
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Tingting Li
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Guoan Gao
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Zhao-Qian Teng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qinmiao Sun
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dahua Chen
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China; Southwest United Graduate School, Kunming 650500, China.
| | - Ying Cheng
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China; Southwest United Graduate School, Kunming 650500, China.
| |
Collapse
|
6
|
Scoyni F, Giudice L, Väänänen M, Downes N, Korhonen P, Choo XY, Välimäki N, Mäkinen P, Korvenlaita N, Rozemuller AJ, de Vries HE, Polo J, Turunen TA, Ylä‐Herttuala S, Hansen TB, Grubman A, Kaikkonen MU, Malm T. Alzheimer's disease-induced phagocytic microglia express a specific profile of coding and non-coding RNAs. Alzheimers Dement 2024; 20:954-974. [PMID: 37828821 PMCID: PMC10916983 DOI: 10.1002/alz.13502] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disease and the main cause of dementia in the elderly. AD pathology is characterized by accumulation of microglia around the beta-amyloid (Aβ) plaques which assumes disease-specific transcriptional signatures, as for the disease-associated microglia (DAM). However, the regulators of microglial phagocytosis are still unknown. METHODS We isolated Aβ-laden microglia from the brain of 5xFAD mice for RNA sequencing to characterize the transcriptional signature in phagocytic microglia and to identify the key non-coding RNAs capable of regulating microglial phagocytosis. Through spatial sequencing, we show the transcriptional changes of microglia in the AD mouse brain in relation to Aβ proximity. RESULTS Finally, we show that phagocytic messenger RNAs are regulated by miR-7a-5p, miR-29a-3p and miR-146a-5p microRNAs and segregate the DAM population into phagocytic and non-phagocytic states. DISCUSSION Our study pinpoints key regulators of microglial Aβ clearing capacity suggesting new targets for future therapeutic approaches.
Collapse
Affiliation(s)
- Flavia Scoyni
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Luca Giudice
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Mari‐Anna Väänänen
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Nicholas Downes
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Paula Korhonen
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Xin Yi Choo
- Department of Anatomy and Developmental BiologyMonash UniversityClaytonAustralia
- Development and Stem Cells ProgramMonash Biomedicine Discovery InstituteClaytonVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| | - Nelli‐Noora Välimäki
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Petri Mäkinen
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Nea Korvenlaita
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Annemieke J Rozemuller
- Department of Pathology, Amsterdam UMC, Vrije Universiteit AmsterdamVU University Medical CenterAmsterdamMBthe Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit AmsterdamVU University Medical CenterAmsterdamMBthe Netherlands
| | - Jose Polo
- Department of Anatomy and Developmental BiologyMonash UniversityClaytonAustralia
- Development and Stem Cells ProgramMonash Biomedicine Discovery InstituteClaytonVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| | - Tiia A Turunen
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Seppo Ylä‐Herttuala
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Thomas B Hansen
- Interdisciplinary Nanoscience CenterDepartment of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
- Present address:
Targovax ASALysaker1366Norway
| | - Alexandra Grubman
- Department of Anatomy and Developmental BiologyMonash UniversityClaytonAustralia
- Development and Stem Cells ProgramMonash Biomedicine Discovery InstituteClaytonVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| | - Minna U Kaikkonen
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Tarja Malm
- A.I.Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| |
Collapse
|
7
|
Getachew B, Hauser SR, Bennani S, El Kouhen N, Sari Y, Tizabi Y. Adolescent alcohol drinking interaction with the gut microbiome: implications for adult alcohol use disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2024; 4:11881. [PMID: 38322648 PMCID: PMC10846679 DOI: 10.3389/adar.2024.11881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024]
Abstract
Reciprocal communication between the gut microbiota and the brain, commonly referred to as the "gut-brain-axis" is crucial in maintaining overall physiological homeostasis. Gut microbiota development and brain maturation (neuronal connectivity and plasticity) appear to be synchronized and to follow the same timeline during childhood (immature), adolescence (expansion) and adulthood (completion). It is important to note that the mesolimbic reward circuitry develops early on, whereas the maturation of the inhibitory frontal cortical neurons is delayed. This imbalance can lead to increased acquirement of reward-seeking and risk-taking behaviors during adolescence, and consequently eventuate in heightened risk for substance abuse. Thus, there is high initiation of alcohol drinking in early adolescence that significantly increases the risk of alcohol use disorder (AUD) in adulthood. The underlying causes for heightened AUD risk are not well understood. It is suggested that alcohol-associated gut microbiota impairment during adolescence plays a key role in AUD neurodevelopment in adulthood. Furthermore, alcohol-induced dysregulation of microglia, either directly or indirectly through interaction with gut microbiota, may be a critical neuroinflammatory pathway leading to neurodevelopmental impairments and AUD. In this review article, we highlight the influence of adolescent alcohol drinking on gut microbiota, gut-brain axis and microglia, and eventual manifestation of AUD. Furthermore, novel therapeutic interventions via gut microbiota manipulations are discussed briefly.
Collapse
Affiliation(s)
- Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Sheketha R. Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
8
|
Frumer GR, Shin SH, Jung S, Kim JS. Not just Glia-Dissecting brain macrophages in the mouse. Glia 2024; 72:5-18. [PMID: 37501579 DOI: 10.1002/glia.24445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Macrophages have emerged as critical cellular components of the central nervous system (CNS), promoting development, maintenance, and immune defense of the CNS. Here we will review recent advances in our understanding of brain macrophage heterogeneity, including microglia and border-associated macrophages, focusing on the mouse. Emphasis will be given to the discussion of strengths and limitations of the experimental approaches that have led to the recent insights and hold promise to further deepen our mechanistic understanding of brain macrophages that might eventually allow to harness their activities for the management of CNS pathologies.
Collapse
Affiliation(s)
- Gal Ronit Frumer
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sun-Hye Shin
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Steffen Jung
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jung-Seok Kim
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
9
|
Kim JS, Jung S. Visualization, Fate Mapping, Ablation, and Mutagenesis of Microglia in the Mouse Brain. ADVANCES IN NEUROBIOLOGY 2024; 37:53-63. [PMID: 39207686 DOI: 10.1007/978-3-031-55529-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Since the classical studies of Pío del Río-Hortega, microglia research has come a long way. In particular, recent advances in bulk and single-cell (sc) transcriptomics have yielded many fascinating new insights into these intriguing immune cells at the interface with the central nervous system (CNS), both in small animal models and human samples. In parallel, tools developed by advanced mouse genetics have revealed the unique ontogeny of microglia and their striking dynamic interactions with other cells in the brain parenchyma. In this chapter, we will discuss various applications of the Cre/loxP-based approach that have enabled the study of microglia in their physiological context of the mouse brain. We will highlight selected key findings that have shaped our current understanding of these cells and discuss the technical intricacies of the Cre/loxP approach and some remaining challenges.
Collapse
Affiliation(s)
- Jung-Seok Kim
- Department of Immunology and Regenerative Biology (IRB), Weizmann Institute of Science, Rehovot, Israel.
| | - Steffen Jung
- Department of Immunology and Regenerative Biology (IRB), Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
10
|
Tripathi A, Rai N, Perles A, Jones C, Dutta R. Dicer deficiency in microglia leads to accelerated demyelination and failed remyelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562812. [PMID: 37905110 PMCID: PMC10614879 DOI: 10.1101/2023.10.17.562812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and are important regulators of normal brain functions. In CNS demyelinating diseases like multiple sclerosis (MS), the functions of these cells are of particular interest. Here we probed the impact of microRNA (miRNA)-mediated post-transcriptional gene regulation using a mouse model lacking microglia/macrophage-specific Dicer expression during demyelination and remyelination. Conditional Dicer ablation and loss of miRNAs in adult microglia led to extensive demyelination and impaired myelin processing. Interestingly, demyelination was accompanied by increased apoptosis of mature oligodendrocytes (OLs) and arresting OL progenitor cells (OPCs) in the precursor stage. At the transcriptional level, Dicer -deficient microglia led to downregulation of microglial homeostatic genes, increased cell proliferation, and a shift towards a disease-associated phenotype. Loss of remyelination efficiency in these mice was accompanied by stalling of OPCs in the precursor stage. Collectively, these results highlight a new role of microglial miRNAs in promoting a pro-regenerative phenotype in addition to promoting OPC maturation and differentiation during demyelination and remyelination.
Collapse
|
11
|
Diebold M, Fehrenbacher L, Frosch M, Prinz M. How myeloid cells shape experimental autoimmune encephalomyelitis: At the crossroads of outside-in immunity. Eur J Immunol 2023; 53:e2250234. [PMID: 37505465 DOI: 10.1002/eji.202250234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/21/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of central nervous system (CNS) autoimmunity. It is most commonly used to mimic aspects of multiple sclerosis (MS), a demyelinating disorder of the human brain and spinal cord. The innate immune response displays one of the core pathophysiological features linked to both the acute and chronic stages of MS. Hence, understanding and targeting the innate immune response is essential. Microglia and other CNS resident MUs, as well as infiltrating myeloid cells, diverge substantially in terms of both their biology and their roles in EAE. Recent advances in the field show that antigen presentation, as well as disease-propagating and regulatory interactions with lymphocytes, can be attributed to specific myeloid cell types and cell states in EAE lesions, following a distinct temporal pattern during disease initiation, propagation and recovery. Furthermore, single-cell techniques enable the assessment of characteristic proinflammatory as well as beneficial cell states, and identification of potential treatment targets. Here, we discuss the principles of EAE induction and protocols for varying experimental paradigms, the composition of the myeloid compartment of the CNS during health and disease, and systematically review effects on myeloid cells for therapeutic approaches in EAE.
Collapse
Affiliation(s)
- Martin Diebold
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Luca Fehrenbacher
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Maximilian Frosch
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Walsh AD, Stone S, Freytag S, Aprico A, Kilpatrick TJ, Ansell BRE, Binder MD. Mouse microglia express unique miRNA-mRNA networks to facilitate age-specific functions in the developing central nervous system. Commun Biol 2023; 6:555. [PMID: 37217597 DOI: 10.1038/s42003-023-04926-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
Microglia regulate multiple processes in the central nervous system, exhibiting a considerable level of cellular plasticity which is facilitated by an equally dynamic transcriptional environment. While many gene networks that regulate microglial functions have been characterised, the influence of epigenetic regulators such as small non-coding microRNAs (miRNAs) is less well defined. We have sequenced the miRNAome and mRNAome of mouse microglia during brain development and adult homeostasis, identifying unique profiles of known and novel miRNAs. Microglia express both a consistently enriched miRNA signature as well as temporally distinctive subsets of miRNAs. We generated robust miRNA-mRNA networks related to fundamental developmental processes, in addition to networks associated with immune function and dysregulated disease states. There was no apparent influence of sex on miRNA expression. This study reveals a unique developmental trajectory of miRNA expression in microglia during critical stages of CNS development, establishing miRNAs as important modulators of microglial phenotype.
Collapse
Affiliation(s)
- Alexander D Walsh
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC, 3052, Australia
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sarrabeth Stone
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC, 3052, Australia
| | - Saskia Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Andrea Aprico
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC, 3052, Australia
| | - Trevor J Kilpatrick
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC, 3052, Australia
| | - Brendan R E Ansell
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Michele D Binder
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
13
|
Liu Q, Huang Y, Duan M, Yang Q, Ren B, Tang F. Microglia as Therapeutic Target for Radiation-Induced Brain Injury. Int J Mol Sci 2022; 23:8286. [PMID: 35955439 PMCID: PMC9368164 DOI: 10.3390/ijms23158286] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Radiation-induced brain injury (RIBI) after radiotherapy has become an increasingly important factor affecting the prognosis of patients with head and neck tumor. With the delivery of high doses of radiation to brain tissue, microglia rapidly transit to a pro-inflammatory phenotype, upregulate phagocytic machinery, and reduce the release of neurotrophic factors. Persistently activated microglia mediate the progression of chronic neuroinflammation, which may inhibit brain neurogenesis leading to the occurrence of neurocognitive disorders at the advanced stage of RIBI. Fully understanding the microglial pathophysiology and cellular and molecular mechanisms after irradiation may facilitate the development of novel therapy by targeting microglia to prevent RIBI and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qun Liu
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Yan Huang
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Mengyun Duan
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Qun Yang
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Boxu Ren
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
14
|
Mehl LC, Manjally AV, Bouadi O, Gibson EM, Tay TL. Microglia in brain development and regeneration. Development 2022; 149:275253. [PMID: 35502782 PMCID: PMC9124570 DOI: 10.1242/dev.200425] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It has recently emerged that microglia, the tissue-resident macrophages of the central nervous system, play significant non-innate immune roles to support the development, maintenance, homeostasis and repair of the brain. Apart from being highly specialized brain phagocytes, microglia modulate the development and functions of neurons and glial cells through both direct and indirect interactions. Thus, recognizing the elements that influence the homeostasis and heterogeneity of microglia in normal brain development is crucial to understanding the mechanisms that lead to early disease pathogenesis of neurodevelopmental disorders. In this Review, we discuss recent studies that have elucidated the physiological development of microglia and summarize our knowledge of their non-innate immune functions in brain development and tissue repair.
Collapse
Affiliation(s)
- Lindsey C Mehl
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | | | - Ouzéna Bouadi
- Faculty of Biology, University of Freiburg, Freiburg, 79104, Germany
| | - Erin M Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Tuan L Tay
- Faculty of Biology, University of Freiburg, Freiburg, 79104, Germany.,BrainLinks-BrainTools Centre, University of Freiburg, Freiburg, 79110, Germany.,Freiburg Institute of Advanced Studies, University of Freiburg, Freiburg, 79104, Germany
| |
Collapse
|
15
|
Yang Y, Guo L, Wang J, Li W, Zhou X, Zhang C, Han C. Arglabin regulates microglia polarization to relieve neuroinflammation in Alzheimer's disease. J Biochem Mol Toxicol 2022; 36:e23045. [PMID: 35289014 DOI: 10.1002/jbt.23045] [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: 12/23/2021] [Revised: 01/29/2022] [Accepted: 03/02/2022] [Indexed: 11/08/2022]
Abstract
Arglabin (Arg) is a derivative of parthenolide. At present, there are few reports on the pharmacological effects and targets of Arg. In this study, we aimed to explore the relationship between Arg and NF-κB (P50) and the intervention effects of Arg on neuroinflammation. BV2 cells were cultured in vitro. LPS/IFN-γ was used to induce M1 polarization. After Arg intervention, the cytokine expression of M1 and M2 cell marker was detected, the expression of CD86 was detected by immunofluorescence (IF) staining, the levels of P50 and p-P50 were detected by Western blot and the expression of ROS was by DCFH-DA. AfterP50 knockout, we investigated the effect of P50 on the polarization of BV2 cells. Four-month-old APP/PS1 (AD) mice were treated with Arg by intragastric administration, followed by detection of the expression of CD86, CD206, and IBA-1 by IF staining, Finally, molecular-protein docking and Pull-down assays were used to validate the targeted binding relationship between P50 and Arg. Arg could inhibit the M1 polarization of BV2 cells, decrease the levels of TNF-α, IL-1β, IL-6, iNOS, and IL-12, and simultaneously inhibit the expression of P50 and p-P50. P50 knockout could inhibit the M1 polarization of BV2 cells, and P50 played an important role in the polarization of BV2 cells. Molecular docking and pull-down assays revealed that Arg and P50 had a targeted binding relationship. Animal experiments showed that Arg could regulate the polarization level of M1-M2 cells, increase the proportion of M2 cells, decrease the degree of nerve injury and suppress the expression of P50 and p-P50. In this study, we found that Arg could target P50 to regulate reprogramming of BV2 cells, inhibit M1 polarization, and increase the level of M2 cells, thereby exerting a neuroprotective effect.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - Li Guo
- Department of Center Laboratory, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - Jin Wang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - WenYan Li
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - Xiaohong Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - Caiqun Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| | - Chenyang Han
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Hangzhou, China
| |
Collapse
|
16
|
Pan TT, Gao W, Song ZH, Long DD, Cao P, Hu R, Chen DY, Zhou WJ, Jin Y, Hu SS, Wei W, Chai XQ, Zhang Z, Wang D. Glutamatergic neurons and myeloid cells in the anterior cingulate cortex mediate secondary hyperalgesia in chronic joint inflammatory pain. Brain Behav Immun 2022; 101:62-77. [PMID: 34973395 DOI: 10.1016/j.bbi.2021.12.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Affiliation(s)
- Ting-Ting Pan
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Wei Gao
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zi-Hua Song
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China; Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Dan-Dan Long
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Peng Cao
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Rui Hu
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Dan-Yang Chen
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Wen-Jie Zhou
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Yan Jin
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Shan-Shan Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Xiao-Qing Chai
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhi Zhang
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Di Wang
- Pain Clinic, Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
17
|
Cuadros MA, Sepulveda MR, Martin-Oliva D, Marín-Teva JL, Neubrand VE. Microglia and Microglia-Like Cells: Similar but Different. Front Cell Neurosci 2022; 16:816439. [PMID: 35197828 PMCID: PMC8859783 DOI: 10.3389/fncel.2022.816439] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Microglia are the tissue-resident macrophages of the central nervous parenchyma. In mammals, microglia are thought to originate from yolk sac precursors and posteriorly maintained through the entire life of the organism. However, the contribution of microglial cells from other sources should also be considered. In addition to “true” or “bona-fide” microglia, which are of embryonic origin, the so-called “microglia-like cells” are hematopoietic cells of bone marrow origin that can engraft the mature brain mainly under pathological conditions. These cells implement great parts of the microglial immune phenotype, but they do not completely adopt the “true microglia” features. Because of their pronounced similarity, true microglia and microglia-like cells are usually considered together as one population. In this review, we discuss the origin and development of these two distinct cell types and their differences. We will also review the factors determining the appearance and presence of microglia-like cells, which can vary among species. This knowledge might contribute to the development of therapeutic strategies aiming at microglial cells for the treatment of diseases in which they are involved, for example neurodegenerative disorders like Alzheimer’s and Parkinson’s diseases.
Collapse
Affiliation(s)
- Miguel A Cuadros
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - M Rosario Sepulveda
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - David Martin-Oliva
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - José L Marín-Teva
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - Veronika E Neubrand
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| |
Collapse
|
18
|
Peeples ES, Sahar NE, Snyder W, Mirnics K. Early Brain microRNA/mRNA Expression is Region-Specific After Neonatal Hypoxic-Ischemic Injury in a Mouse Model. Front Genet 2022; 13:841043. [PMID: 35251138 PMCID: PMC8890746 DOI: 10.3389/fgene.2022.841043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: MicroRNAs (miRNAs) may be promising therapeutic targets for neonatal hypoxic-ischemic brain injury (HIBI) but targeting miRNA-based therapy will require more precise understanding of endogenous brain miRNA expression. Methods: Postnatal day 9 mouse pups underwent HIBI by unilateral carotid ligation + hypoxia or sham surgery. Next-generation miRNA sequencing and mRNA Neuroinflammation panels were performed on ipsilateral cortex, striatum/thalamus, and cerebellum of each group at 30 min after injury. Targeted canonical pathways were predicted by KEGG analysis. Results: Sixty-one unique miRNAs showed differential expression (DE) in at least one region; nine in more than one region, including miR-410-5p, -1264-3p, 1298-5p, -5,126, and -34b-3p. Forty-four mRNAs showed DE in at least one region; 16 in more than one region. MiRNAs showing DE primarily targeted metabolic pathways, while mRNAs targeted inflammatory and cell death pathways. Minimal miRNA-mRNA interactions were seen at 30 min after HIBI. Conclusion: This study identified miRNAs that deserve future study to assess their potential as therapeutic targets in neonatal HIBI. Additionally, the differences in miRNA expression between regions suggest that future studies assessing brain miRNA expression to guide therapy development should consider evaluating individual brain regions rather than whole brain to ensure the sensitivity needed for the development of targeted therapies.
Collapse
Affiliation(s)
- Eric S. Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pediatrics, Children’s Hospital & Medical Center, Omaha, NE, United States
- Child Health Research Institute, Omaha, NE, United States
| | - Namood-e Sahar
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
- Child Health Research Institute, Omaha, NE, United States
| | - William Snyder
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
- Child Health Research Institute, Omaha, NE, United States
| | - Karoly Mirnics
- Child Health Research Institute, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
19
|
Li L, Qi C, Liu Y, Shen Y, Zhao X, Qin H, Zhang Y, Yu T. MicroRNA miR-27b-3p regulate microglial inflammation response and cell apoptosis by inhibiting A20 (TNF-α-induced protein 3). Bioengineered 2021; 12:9902-9913. [PMID: 34895052 PMCID: PMC8810141 DOI: 10.1080/21655979.2021.1969195] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inflammatory reaction exerts a pivotal role in secondary damage after cerebral hemorrhage and spinal cord injury. miRNAs can both promote and inhibit inflammatory actions among microglial cells. The objective of the present paper was to figure out whether miR-27b-3p produced regulatory effects during processes of microglial inflammation. Lipopolysaccharides (LPS) were used to prepare microglial activation models. Following miR-27b-3p overexpression and interference, the RNA and protein levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β were subjected to real-time fluorescent quantitative PCR (qPCR) and western blot assays, respectively. Cellular apoptosis was subjected to flow cytometry and miR-27b-3p target genes were visualized using a dual luciferase reporter system for verification. The levels of TNF-α, IL-6, and IL-1β mRNA in miR-27b-3p-overexpressed microglial cells were markedly increased compared to the control. Apoptosis of microglial cells was increased markedly in the overexpressed miR-27b-3p group compared to the negative control. Conversely, a different result was presented in the microglial transfected with miR-27b-3p inhibitors. The downregulation of A20, a miR-27b-3p target gene, mediated levels of TNF-α, IL-6, and IL-1β. Furthermore, A20 reduced microglial apoptosis. These data revealed that miR-27b-3p could mediate not only microglia activation but also neuroinflammation via downregulating A20 expression. Thus, miR-27b-3p is regarded as gene therapy in treating cerebral hemorrhage and spinal cord injury.
Collapse
Affiliation(s)
- Liping Li
- Department of Orthopedic Surgery, The Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Orthopedic Surgery, The Second Clinical Medical College of Qingdao University, Qingdao, Shandong, China
| | - Chao Qi
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yuanyuan Liu
- Department of Oncology, The Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Oncology, The Second Clinical Medical College of Qingdao University, Qingdao, Shandong, China
| | - Youliang Shen
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xia Zhao
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Han Qin
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yi Zhang
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tengbo Yu
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
20
|
Bokobza C, Joshi P, Schang AL, Csaba Z, Faivre V, Montané A, Galland A, Benmamar-Badel A, Bosher E, Lebon S, Schwendimann L, Mani S, Dournaud P, Besson V, Fleiss B, Gressens P, Van Steenwinckel J. miR-146b Protects the Perinatal Brain against Microglia-Induced Hypomyelination. Ann Neurol 2021; 91:48-65. [PMID: 34741343 PMCID: PMC9298799 DOI: 10.1002/ana.26263] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022]
Abstract
Objectives In the premature newborn, perinatal inflammation mediated by microglia contributes significantly to neurodevelopmental injuries including white matter injury (WMI). Brain inflammation alters development through neuroinflammatory processes mediated by activation of homeostatic microglia toward a pro‐inflammatory and neurotoxic phenotype. Investigating immune regulators of microglial activation is crucial to find effective strategies to prevent and treat WMI. Methods Ex vivo microglial cultures and a mouse model of WMI induced by perinatal inflammation (interleukin‐1‐beta [IL‐1β] and postnatal days 1–5) were used to uncover and elucidate the role of microRNA‐146b‐5p in microglial activation and WMI. Results A specific reduction in vivo in microglia of Dicer, a protein required for microRNAs maturation, reduces pro‐inflammatory activation of microglia and prevents hypomyelination in our model of WMI. Microglial miRNome analysis in the WMI model identified miRNA‐146b‐5p as a candidate modulator of microglial activation. Ex vivo microglial cell culture treated with the pro‐inflammatory stimulus lipopolysaccharide (LPS) led to overexpression of immunomodulatory miRNA‐146b‐5p but its drastic reduction in the microglial extracellular vesicles (EVs). To increase miRNA‐146b‐5p expression, we used a 3DNA nanocarrier to deliver synthetic miRNA‐146b‐5p specifically to microglia. Enhancing microglial miRNA‐146b‐5p overexpression significantly decreased LPS‐induced activation, downregulated IRAK1, and restored miRNA‐146b‐5p levels in EVs. In our WMI model, 3DNA miRNA‐146b‐5p treatment significantly prevented microglial activation, hypomyelination, and cognitive defect induced by perinatal inflammation. Interpretations These findings support that miRNA‐146b‐5p is a major regulator of microglia phenotype and could be targeted to reduce the incidence and the severity of perinatal brain injuries and their long‐term consequences. ANN NEUROL 2022;91:48–65
Collapse
Affiliation(s)
- Cindy Bokobza
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Pooja Joshi
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Anne-Laure Schang
- Université de Paris, Centre de recherche en Epidémiologie et Statistiques, Inserm, Paris, France
| | - Zsolt Csaba
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Valérie Faivre
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Amélie Montané
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Anne Galland
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | | | | | - Sophie Lebon
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | | | - Shyamala Mani
- Université de Paris, NeuroDiderot, Inserm, Paris, France.,Curadev Pharma, Pvt. Ltd, Noida, India
| | | | - Valerie Besson
- Université de Paris, Faculté de Pharmacie de Paris, UMR-S1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Bobbi Fleiss
- Université de Paris, NeuroDiderot, Inserm, Paris, France.,School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | | | | |
Collapse
|
21
|
Alcohol induced impairment/abnormalities in brain: Role of MicroRNAs. Neurotoxicology 2021; 87:11-23. [PMID: 34478768 DOI: 10.1016/j.neuro.2021.08.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/12/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022]
Abstract
Alcohol is a highly toxic substance and has teratogenic properties that can lead to a wide range of developmental disorders. Excessive use of alcohol can change the structural and functional aspects of developed brain and other organs. Which can further lead to significant health, social and economic implications in many countries of the world. Convincing evidence support the involvement of microRNAs (miRNAs) as important post-transcriptional regulators of gene expression in neurodevelopment and maintenance. They also show differential expression following an injury. MiRNAs are the special class of small non coding RNAs that can modify the gene by targeting the mRNA and fine tune the development of cells to organs. Numerous pieces of evidences have shown the relationship between miRNA, alcohol and brain damage. These studies also show how miRNA controls different cellular mechanisms involved in the development of alcohol use disorder. With the increasing number of research studies, the roles of miRNAs following alcohol-induced injury could help researchers to recognize alternative therapeutic methods to treat/cure alcohol-induced brain damage. The present review summarizes the available data and brings together the important miRNAs, that play a crucial role in alcohol-induced brain damage, which will help in better understanding complex mechanisms. Identifying these miRNAs will not only expand the current knowledge but can lead to the identification of better targets for the development of novel therapeutic interventions.
Collapse
|
22
|
Rayasam A, Fukuzaki Y, Vexler ZS. Microglia-leucocyte axis in cerebral ischaemia and inflammation in the developing brain. Acta Physiol (Oxf) 2021; 233:e13674. [PMID: 33991400 DOI: 10.1111/apha.13674] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022]
Abstract
Development of the Central Nervous System (CNS) is reliant on the proper function of numerous intricately orchestrated mechanisms that mature independently, including constant communication between the CNS and the peripheral immune system. This review summarizes experimental knowledge of how cerebral ischaemia in infants and children alters physiological communication between leucocytes, brain immune cells, microglia and the neurovascular unit (NVU)-the "microglia-leucocyte axis"-and contributes to acute and long-term brain injury. We outline physiological development of CNS barriers in relation to microglial and leucocyte maturation and the plethora of mechanisms by which microglia and peripheral leucocytes communicate during postnatal period, including receptor-mediated and intracellular inflammatory signalling, lipids, soluble factors and extracellular vesicles. We focus on the "microglia-leucocyte axis" in rodent models of most common ischaemic brain diseases in the at-term infants, hypoxic-ischaemic encephalopathy (HIE) and focal arterial stroke and discuss commonalities and distinctions of immune-neurovascular mechanisms in neonatal and childhood stroke compared to stroke in adults. Given that hypoxic and ischaemic brain damage involve Toll-like receptor (TLR) activation, we discuss the modulatory role of viral and bacterial TLR2/3/4-mediated infection in HIE, perinatal and childhood stroke. Furthermore, we provide perspective of the dynamics and contribution of the axis in cerebral ischaemia depending on the CNS maturational stage at the time of insult, and modulation independently and in consort by individual axis components and in a sex dependent ways. Improved understanding on how to modify crosstalk between microglia and leucocytes will aid in developing age-appropriate therapies for infants and children who suffered cerebral ischaemia.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Neurology University of California San Francisco San Francisco CA USA
| | - Yumi Fukuzaki
- Department of Neurology University of California San Francisco San Francisco CA USA
| | - Zinaida S. Vexler
- Department of Neurology University of California San Francisco San Francisco CA USA
| |
Collapse
|
23
|
Traetta ME, Uccelli NA, Zárate SC, Gómez Cuautle D, Ramos AJ, Reinés A. Long-Lasting Changes in Glial Cells Isolated From Rats Subjected to the Valproic Acid Model of Autism Spectrum Disorder. Front Pharmacol 2021; 12:707859. [PMID: 34421599 PMCID: PMC8374432 DOI: 10.3389/fphar.2021.707859] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023] Open
Abstract
Synaptic alterations concomitant with neuroinflammation have been described in patients and experimental models of autism spectrum disorder (ASD). However, the role of microglia and astroglia in relation to synaptic changes is poorly understood. Male Wistar rats prenatally exposed to valproic acid (VPA, 450 mg/kg, i.p.) or saline (control) at embryonic day 10.5 were used to study synapses, microglia, and astroglia in the prefrontal cortex (PFC) at postnatal days 3 and 35 (PND3 and PND35). Primary cultures of cortical neurons, microglia, and astroglia isolated from control and VPA animals were used to study each cell type individually, neuron-microglia and microglia-astroglia crosstalk. In the PFC of VPA rats, synaptic changes characterized by an increase in the number of excitatory synapses were evidenced at PND3 and persisted until PND35. At PND3, microglia and astroglia from VPA animals were morphologically similar to those of age-matched controls, whereas at PND35, reactive microgliosis and astrogliosis were observed in the PFC of VPA animals. Cortical neurons isolated from VPA rats mimicked in vitro the synaptic pattern seen in vivo. Cortical microglia and astroglia isolated from VPA animals exhibited reactive morphology, increased pro-inflammatory cytokines, and a compromised miRNA processing machinery. Microglia from VPA animals also showed resistance to a phagocytic challenge. In the presence of neurons from VPA animals, microglia isolated from VPA rats revealed a non-reactive morphology and promoted neurite outgrowth, while microglia from control animals displayed a reactive profile and promoted dendritic retraction. In microglia-astroglia co-cultures, microglia from VPA animals displayed a reactive profile and exacerbated astrocyte reactivity. Our study indicates that cortical microglia from VPA animals are insensitive or adapted to neuronal cues expressed by neurons from VPA animals. Further, long-term in vivo microgliosis could be the result of altered microglia-astroglia crosstalk in VPA animals. Thus, our study highlights cortical microglia-astroglia communication as a new mechanism implicated in neuroinflammation in ASD; consequently, we propose that this crosstalk is a potential target for interventions in this disorder.
Collapse
Affiliation(s)
- Marianela Evelyn Traetta
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nonthué Alejandra Uccelli
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sandra Cristina Zárate
- Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas (INBIOMED), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante Gómez Cuautle
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Medicina, Departamento de Histología, Embriología, Biología Celular y Genética, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Reinés
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), CONICET - Universidad de Buenos Aires, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
He CQ, Mao L, Yao J, Zhao WC, Huang B, Hu N, Long DX. The Threshold Effects of Low-Dose-Rate Radiation on miRNA-Mediated Neurodevelopment of Zebrafish. Radiat Res 2021; 196:633-646. [PMID: 34399425 DOI: 10.1667/rade-20-00265.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 07/29/2021] [Indexed: 11/03/2022]
Abstract
The biological effects and regulatory mechanisms of low-dose and low-dose-rate radiation are still rather controversial. Therefore, in this study we investigated the effects of low-dose-rate radiation on zebrafish neurodevelopment and the role of miRNAs in radiation-induced neurodevelopment. Zebrafish embryos received prolonged gamma-ray irradiation (0 mGy/h, 0.1 mGy/h, 0.2 mGy/h, 0.4 mGy/h) during development. Neurodevelopmental indicators included mortality, malformation rate, swimming speed, as well as the morphology changes of the lateral line system and brain tissue. Additionally, spatiotemporal expression of development-related miRNAs (dre-miR-196a-5p, dre-miR-210-3p, dre-miR-338) and miRNA processing enzymes genes (Dicer and Drosha) were assessed by qRT-PCR and whole mount in situ hybridization (WISH). The results revealed a decline in mortality, malformation and swimming speed, with normal histological and morphological appearance, in zebrafish that received 0.1 mGy/h; however, increased mortality, malformation and swimming speed were observed, with pathological changes, in zebrafish that received 0.2 mGy/h and 0.4 mGy/h. The expression of miRNA processing enzyme genes was altered after irradiation, and miRNAs expression was downregulated in the 0.1 mGy/h group, and upregulated in the 0.2 mGy/h and 0.4 mGy/h groups. Furthermore, ectopic expression of dre-miR-210-3p, Dicer and Drosha was also observed in the 0.4 mGy/h group. In conclusion, the effect of low-dose and low-dose-rate radiation on neurodevelopment follows the threshold model, under the regulation of miRNAs, excitatory effects occurred at a dose rate of 0.1 mGy/h and toxic effects occurred at a dose rate of 0.2 mGy/h and 0.4 mGy/h.
Collapse
Affiliation(s)
- Chu-Qi He
- School of Public Health, University of South China, Hengyang 421001, PR China.,Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang, Hunan 421001, China
| | - Liang Mao
- School of Public Health, University of South China, Hengyang 421001, PR China.,Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang, Hunan 421001, China
| | - Jin Yao
- School of Public Health, University of South China, Hengyang 421001, PR China.,Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang, Hunan 421001, China
| | - Wei-Chao Zhao
- School of Public Health, University of South China, Hengyang 421001, PR China.,Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang, Hunan 421001, China
| | - Bo Huang
- School of Public Health, University of South China, Hengyang 421001, PR China.,Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang, Hunan 421001, China
| | - Nan Hu
- Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, China
| | - Ding-Xin Long
- School of Public Health, University of South China, Hengyang 421001, PR China.,Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, Hengyang, Hunan 421001, China
| |
Collapse
|
25
|
Kashif H, Shah D, Sukumari-Ramesh S. Dysregulation of microRNA and Intracerebral Hemorrhage: Roles in Neuroinflammation. Int J Mol Sci 2021; 22:8115. [PMID: 34360881 PMCID: PMC8347974 DOI: 10.3390/ijms22158115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/23/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a major public health problem and devastating subtype of stroke with high morbidity and mortality. Notably, there is no effective treatment for ICH. Neuroinflammation, a pathological hallmark of ICH, contributes to both brain injury and repair and hence, it is regarded as a potential target for therapeutic intervention. Recent studies document that microRNAs, small non-coding RNA molecules, can regulate inflammatory brain response after ICH and are viable molecular targets to alter brain function. Therefore, there is an escalating interest in studying the role of microRNAs in the pathophysiology of ICH. Herein, we provide, for the first time, an overview of the microRNAs that play roles in ICH-induced neuroinflammation and identify the critical knowledge gap in the field, as it would help design future studies.
Collapse
Affiliation(s)
| | | | - Sangeetha Sukumari-Ramesh
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.K.); (D.S.)
| |
Collapse
|
26
|
Luo X, Jean-Toussaint R, Sacan A, Ajit SK. Differential RNA packaging into small extracellular vesicles by neurons and astrocytes. Cell Commun Signal 2021; 19:75. [PMID: 34246289 PMCID: PMC8272329 DOI: 10.1186/s12964-021-00757-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 06/03/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Small extracellular vesicles (sEVs) mediate intercellular communication by transferring RNA, proteins, and lipids to recipient cells. These cargo molecules are selectively loaded into sEVs and mirror the physiological state of the donor cells. Given that sEVs can cross the blood-brain barrier and their composition can change in neurological disorders, the molecular signatures of sEVs in circulation can be potential disease biomarkers. Characterizing the molecular composition of sEVs from different cell types is an important first step in determining which donor cells contribute to the circulating sEVs. METHODS Cell culture supernatants from primary mouse cortical neurons and astrocytes were used to purify sEVs by differential ultracentrifugation and sEVs were characterized using nanoparticle tracking analysis, transmission electron microscopy and western blot. RNA sequencing was used to determine differential expression and loading patterns of miRNAs in sEVs released by primary neurons and astrocytes. Motif analysis was conducted on enriched miRNAs in sEVs and their respective donor cells. RESULTS Sequencing total cellular RNA, and miRNAs from sEVs isolated from culture media of postnatal mouse cortical neurons and astrocytes revealed a distinct profile between sEVs and their corresponding cells. Though the total number of detected miRNAs in astrocytes was greater than neurons, neurons expressed more sEV-associated miRNAs than astrocytes. Only 20.7% of astrocytic miRNAs were loaded into sEVs, while 41.0% of neuronal miRNAs were loaded into sEVs, suggesting differences in the cellular sorting mechanisms. We identified short RNA sequence motifs, or EXOmotifs, on the miRNAs that were differentially loaded or excluded from sEVs. A sequence motif GUAC was enriched in astrocytic sEVs. miRNAs preferably retained in neurons or astrocytes had a similar RNA motif CACACA, suggesting a cell-type-independent mechanism to maintain cellular miRNAs. mRNAs of five RNA-binding proteins associated with passive or active RNA sorting into sEVs were differentially expressed between neurons and astrocytes, one of which, major vault protein was higher in astrocytes than in neurons and detected in astrocytic sEVs. CONCLUSIONS Our studies suggest differences in RNA sorting into sEVs. These differences in miRNA signatures can be used for determining the cellular sources of sEVs altered in neurological disorders. Video abstract.
Collapse
Affiliation(s)
- Xuan Luo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102 USA
| | - Renée Jean-Toussaint
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102 USA
| | - Ahmet Sacan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104 USA
| | - Seena K. Ajit
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102 USA
| |
Collapse
|
27
|
Suster I, Feng Y. Multifaceted Regulation of MicroRNA Biogenesis: Essential Roles and Functional Integration in Neuronal and Glial Development. Int J Mol Sci 2021; 22:ijms22136765. [PMID: 34201807 PMCID: PMC8269442 DOI: 10.3390/ijms22136765] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs that function as endogenous gene silencers. Soon after the discovery of miRNAs, a subset of brain-enriched and brain-specific miRNAs were identified and significant advancements were made in delineating miRNA function in brain development. However, understanding the molecular mechanisms that regulate miRNA biogenesis in normal and diseased brains has become a prevailing challenge. Besides transcriptional regulation of miRNA host genes, miRNA processing intermediates are subjected to multifaceted regulation by canonical miRNA processing enzymes, RNA binding proteins (RBPs) and epitranscriptomic modifications. Further still, miRNA activity can be regulated by the sponging activity of other non-coding RNA classes, namely circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs). Differential abundance of these factors in neuronal and glial lineages partly underlies the spatiotemporal expression and function of lineage-specific miRNAs. Here, we review the continuously evolving understanding of the regulation of neuronal and glial miRNA biogenesis at the transcriptional and posttranscriptional levels and the cooperativity of miRNA species in targeting key mRNAs to drive lineage-specific development. In addition, we review dysregulation of neuronal and glial miRNAs and the detrimental impacts which contribute to developmental brain disorders.
Collapse
Affiliation(s)
| | - Yue Feng
- Correspondence: ; Tel.: +1-404-727-0351
| |
Collapse
|
28
|
Dicer Deletion in Astrocytes Inhibits Oligodendroglial Differentiation and Myelination. Neurosci Bull 2021; 37:1135-1146. [PMID: 34106403 PMCID: PMC8353046 DOI: 10.1007/s12264-021-00705-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/13/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence has shown that astrocytes are implicated in regulating oligodendrocyte myelination, but the underlying mechanisms remain largely unknown. To understand whether microRNAs in astrocytes function in regulating oligodendroglial differentiation and myelination in the developing and adult CNS, we generated inducible astrocyte-specific Dicer conditional knockout mice (hGFAP-CreERT; Dicer fl/fl). By using a reporter mouse line (mT/mG), we confirmed that hGFAP-CreERT drives an efficient and astrocyte-specific recombination in the developing CNS, upon tamoxifen treatment from postnatal day 3 (P3) to P7. The Dicer deletion in astrocytes resulted in inhibited oligodendroglial differentiation and myelination in the developing CNS of Dicer cKO mice at P10 and P14, and did not alter the densities of neurons or axons, indicating that Dicer in astrocytes is required for oligodendrocyte myelination. Consequently, the Dicer deletion in astrocytes at P3 resulted in impaired spatial memory and motor coordination at the age of 9 weeks. To understand whether Dicer in astrocytes is also required for remyelination, we induced Dicer deletion in 3-month-old mice and then injected lysolecithin into the corpus callosum to induce demyelination. The Dicer deletion in astrocytes blocked remyelination in the corpus callosum 14 days after induced demyelination. Together, our results indicate that Dicer in astrocytes is required for oligodendroglia myelination in both the developing and adult CNS.
Collapse
|
29
|
Tripathi A, Pandit I, Perles A, Zhou Y, Cheng F, Dutta R. Identifying miRNAs in multiple sclerosis gray matter lesions that correlate with atrophy measures. Ann Clin Transl Neurol 2021; 8:1279-1291. [PMID: 33978322 PMCID: PMC8164853 DOI: 10.1002/acn3.51365] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 03/27/2021] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS). Though MS was initially considered to be a white matter demyelinating disease, myelin loss in cortical gray matter has been reported in all disease stages. We previously identified microRNAs (miRNAs) in white matter lesions (WMLs) that are detected in serum from MS patients. However, miRNA expression profiles in gray matter lesions (GMLs) from progressive MS brains are understudied. METHODS We used a combination of global miRNAs and gene expression profiling of GMLs and independent validation using real-time quantitative polymerase chain reaction (RT-qPCR), immuno-in situ hybridization, and immunohistochemistry. RESULTS Compared to matched myelinated gray matter (GM) regions, we identified 82 miRNAs in GMLs, of which 10 were significantly upregulated and 17 were significantly downregulated. Among these 82 miRNAs, 13 were also detected in serum and importantly were associated with brain atrophy in MS patients. The predicted target mRNAs of these miRNAs belonged to pathways associated with axonal guidance, TGF-β signaling, and FOXO signaling. Further, using state-of-the-art human protein-protein interactome network analysis, we mapped the four key GM atrophy-associated miRNAs (hsa-miR-149*, hsa-miR-20a, hsa-miR-29c, and hsa-miR-25) to their target mRNAs that were also changed in GMLs. INTERPRETATION Our study identifies miRNAs altered in GMLs in progressive MS brains that correlate with atrophy measures. As these miRNAs were also detected in sera of MS patients, these could act as markers of GML demyelination in MS.
Collapse
Affiliation(s)
- Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| | - Ishani Pandit
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Aaron Perles
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yadi Zhou
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Feixiong Cheng
- Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA.,Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
30
|
Zolboot N, Du JX, Zampa F, Lippi G. MicroRNAs Instruct and Maintain Cell Type Diversity in the Nervous System. Front Mol Neurosci 2021; 14:646072. [PMID: 33994943 PMCID: PMC8116551 DOI: 10.3389/fnmol.2021.646072] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Characterizing the diverse cell types that make up the nervous system is essential for understanding how the nervous system is structured and ultimately how it functions. The astonishing range of cellular diversity found in the nervous system emerges from a small pool of neural progenitor cells. These progenitors and their neuronal progeny proceed through sequential gene expression programs to produce different cell lineages and acquire distinct cell fates. These gene expression programs must be tightly regulated in order for the cells to achieve and maintain the proper differentiated state, remain functional throughout life, and avoid cell death. Disruption of developmental programs is associated with a wide range of abnormalities in brain structure and function, further indicating that elucidating their contribution to cellular diversity will be key to understanding brain health. A growing body of evidence suggests that tight regulation of developmental genes requires post-transcriptional regulation of the transcriptome by microRNAs (miRNAs). miRNAs are small non-coding RNAs that function by binding to mRNA targets containing complementary sequences and repressing their translation into protein, thereby providing a layer of precise spatial and temporal control over gene expression. Moreover, the expression profiles and targets of miRNAs show great specificity for distinct cell types, brain regions and developmental stages, suggesting that they are an important parameter of cell type identity. Here, we provide an overview of miRNAs that are critically involved in establishing neural cell identities, focusing on how miRNA-mediated regulation of gene expression modulates neural progenitor expansion, cell fate determination, cell migration, neuronal and glial subtype specification, and finally cell maintenance and survival.
Collapse
Affiliation(s)
- Norjin Zolboot
- The Scripps Research Institute, La Jolla, CA, United States
| | - Jessica X. Du
- The Scripps Research Institute, La Jolla, CA, United States
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Federico Zampa
- The Scripps Research Institute, La Jolla, CA, United States
| | - Giordano Lippi
- The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
31
|
Killy B, Bodendorfer B, Mages J, Ritter K, Schreiber J, Hölscher C, Pracht K, Ekici A, Jäck HM, Lang R. DGCR8 deficiency impairs macrophage growth and unleashes the interferon response to mycobacteria. Life Sci Alliance 2021; 4:4/6/e202000810. [PMID: 33771876 PMCID: PMC8008949 DOI: 10.26508/lsa.202000810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022] Open
Abstract
The mycobacterial cell wall glycolipid trehalose-6,6-dimycolate (TDM) activates macrophages through the C-type lectin receptor MINCLE. Regulation of innate immune cells relies on miRNAs, which may be exploited by mycobacteria to survive and replicate in macrophages. Here, we have used macrophages deficient in the microprocessor component DGCR8 to investigate the impact of miRNA on the response to TDM. Deletion of DGCR8 in bone marrow progenitors reduced macrophage yield, but did not block macrophage differentiation. DGCR8-deficient macrophages showed reduced constitutive and TDM-inducible miRNA expression. RNAseq analysis revealed that they accumulated primary miRNA transcripts and displayed a modest type I IFN signature at baseline. Stimulation with TDM in the absence of DGCR8 induced overshooting expression of IFNβ and IFN-induced genes, which was blocked by antibodies to type I IFN. In contrast, signaling and transcriptional responses to recombinant IFNβ were unaltered. Infection with live Mycobacterium bovis Bacille Calmette-Guerin replicated the enhanced IFN response. Together, our results reveal an essential role for DGCR8 in curbing IFNβ expression macrophage reprogramming by mycobacteria.
Collapse
Affiliation(s)
- Barbara Killy
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Bodendorfer
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Kristina Ritter
- Infection Immunology, Forschungszentrum Borstel, Borstel, Germany
| | - Jonathan Schreiber
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Hölscher
- Infection Immunology, Forschungszentrum Borstel, Borstel, Germany.,German Center for Infection Research (DZIF), Partner Site Borstel, Borstel, Germany
| | - Katharina Pracht
- Division of Molecular Immunology, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arif Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
32
|
Zhang X, Heng Y, Kooistra SM, van Weering HRJ, Brummer ML, Gerrits E, Wesseling EM, Brouwer N, Nijboer TW, Dubbelaar ML, Boddeke EWGM, Eggen BJL. Intrinsic DNA damage repair deficiency results in progressive microglia loss and replacement. Glia 2021; 69:729-745. [PMID: 33068332 PMCID: PMC7821301 DOI: 10.1002/glia.23925] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022]
Abstract
The DNA excision repair protein Ercc1 is important for nucleotide excision, double strand DNA break, and interstrand DNA crosslink repair. In constitutive Ercc1-knockout mice, microglia display increased phagocytosis, proliferation and an enhanced responsiveness to lipopolysaccharide (LPS)-induced peripheral inflammation. However, the intrinsic effects of Ercc1-deficiency on microglia are unclear. In this study, Ercc1 was specifically deleted from Cx3cr1-expressing cells and changes in microglia morphology and immune responses at different times after deletion were determined. Microglia numbers were reduced with approximately 50% at 2-12 months after Ercc1 deletion. Larger and more ramified microglia were observed following Ercc1 deletion both in vivo and in organotypic hippocampal slice cultures. Ercc1-deficient microglia were progressively lost, and during this period, microglia proliferation was transiently increased. Ercc1-deficient microglia were gradually replaced by nondeficient microglia carrying a functional Ercc1 allele. In contrast to constitutive Ercc1-deficient mice, microglia-specific deletion of Ercc1 did not induce microglia activation or increase their responsiveness to a systemic LPS challenge. Gene expression analysis suggested that Ercc1 deletion in microglia induced a transient aging signature, which was different from a priming or disease-associated microglia gene expression profile.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Yang Heng
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Susanne M. Kooistra
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Hilmar R. J. van Weering
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Maaike L. Brummer
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Emma Gerrits
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Evelyn M. Wesseling
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Tjalling W. Nijboer
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Marissa L. Dubbelaar
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Erik W. G. M. Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Center for Healthy Ageing, Department of Cellular and Molecular MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
33
|
Hou J, Zhang J, Cui P, Zhou Y, Liu C, Wu X, Ji Y, Wang S, Cheng B, Ye H, Shu L, Zhang K, Wang D, Xu J, Shu Q, Colonna M, Fang X. TREM2 sustains macrophage-hepatocyte metabolic coordination in nonalcoholic fatty liver disease and sepsis. J Clin Invest 2021; 131:135197. [PMID: 33586673 DOI: 10.1172/jci135197] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
Sepsis is a leading cause of death in critical illness, and its pathophysiology varies depending on preexisting medical conditions. Here we identified nonalcoholic fatty liver disease (NAFLD) as an independent risk factor for sepsis in a large clinical cohort and showed a link between mortality in NAFLD-associated sepsis and hepatic mitochondrial and energetic metabolism dysfunction. Using in vivo and in vitro models of liver lipid overload, we discovered a metabolic coordination between hepatocyte mitochondria and liver macrophages that express triggering receptor expressed on myeloid cells-2 (TREM2). Trem2-deficient macrophages released exosomes that impaired hepatocytic mitochondrial structure and energy supply because of their high content of miR-106b-5p, which blocks Mitofusin 2 (Mfn2). In a mouse model of NAFLD-associated sepsis, TREM2 deficiency accelerated the initial progression of NAFLD and subsequent susceptibility to sepsis. Conversely, overexpression of TREM2 in liver macrophages improved hepatic energy supply and sepsis outcome. This study demonstrates that NAFLD is a risk factor for sepsis, providing a basis for precision treatment, and identifies hepatocyte-macrophage metabolic coordination and TREM2 as potential targets for future clinical trials.
Collapse
Affiliation(s)
- Jinchao Hou
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jue Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ping Cui
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Can Liu
- Department of Thoracic and Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Anesthesiology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Xiaoliang Wu
- Department of Intensive Care Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yun Ji
- Surgical Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sicong Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Baoli Cheng
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Ye
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liqi Shu
- Department of Neurology, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Kai Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiangming Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Prinz M, Masuda T, Wheeler MA, Quintana FJ. Microglia and Central Nervous System-Associated Macrophages-From Origin to Disease Modulation. Annu Rev Immunol 2021; 39:251-277. [PMID: 33556248 DOI: 10.1146/annurev-immunol-093019-110159] [Citation(s) in RCA: 319] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The immune system of the central nervous system (CNS) consists primarily of innate immune cells. These are highly specialized macrophages found either in the parenchyma, called microglia, or at the CNS interfaces, such as leptomeningeal, perivascular, and choroid plexus macrophages. While they were primarily thought of as phagocytes, their function extends well beyond simple removal of cell debris during development and diseases. Brain-resident innate immune cells were found to be plastic, long-lived, and host to an outstanding number of risk genes for multiple pathologies. As a result, they are now considered the most suitable targets for modulating CNS diseases. Additionally, recent single-cell technologies enhanced our molecular understanding of their origins, fates, interactomes, and functional cell statesduring health and perturbation. Here, we review the current state of our understanding and challenges of the myeloid cell biology in the CNS and treatment options for related diseases.
Collapse
Affiliation(s)
- Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, D-79106 Freiburg, Germany; .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, D-79106 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, D-79104 Freiburg, Germany
| | - Takahiro Masuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 812-8582 Fukuoka, Japan;
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA; , .,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA; , .,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
35
|
Walsh AD, Nguyen LT, Binder MD. miRNAs in Microglia: Important Players in Multiple Sclerosis Pathology. ASN Neuro 2021; 13:1759091420981182. [PMID: 33517686 PMCID: PMC7863159 DOI: 10.1177/1759091420981182] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system and important regulators of brain homeostasis. Central to this role is a dynamic phenotypic plasticity that enables microglia to respond to environmental and pathological stimuli. Importantly, different microglial phenotypes can be both beneficial and detrimental to central nervous system health. Chronically activated inflammatory microglia are a hallmark of neurodegeneration, including the autoimmune disease multiple sclerosis (MS). By contrast, microglial phagocytosis of myelin debris is essential for resolving inflammation and promoting remyelination. As such, microglia are being explored as a potential therapeutic target for MS. MicroRNAs (miRNAs) are short non-coding ribonucleic acids that regulate gene expression and act as master regulators of cellular phenotype and function. Dysregulation of certain miRNAs can aberrantly activate and promote specific polarisation states in microglia to modulate their activity in inflammation and neurodegeneration. In addition, miRNA dysregulation is implicated in MS pathogenesis, with circulating biomarkers and lesion specific miRNAs identified as regulators of inflammation and myelination. However, the role of miRNAs in microglia that specifically contribute to MS progression are still largely unknown. miRNAs are being explored as therapeutic agents, providing an opportunity to modulate microglial function in neurodegenerative diseases such as MS. This review will focus firstly on elucidating the complex role of microglia in MS pathogenesis. Secondly, we explore the essential roles of miRNAs in microglial function. Finally, we focus on miRNAs that are implicated in microglial processes that contribute directly to MS pathology, prioritising targets that could inform novel therapeutic approaches to MS.
Collapse
Affiliation(s)
- Alexander D Walsh
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
| | - Linda T Nguyen
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
| | - Michele D Binder
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Melbourne, Australia
| |
Collapse
|
36
|
Saw G, Tang FR. Epigenetic Regulation of the Hippocampus, with Special Reference to Radiation Exposure. Int J Mol Sci 2020; 21:ijms21249514. [PMID: 33327654 PMCID: PMC7765140 DOI: 10.3390/ijms21249514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 01/28/2023] Open
Abstract
The hippocampus is crucial in learning, memory and emotion processing, and is involved in the development of different neurological and neuropsychological disorders. Several epigenetic factors, including DNA methylation, histone modifications and non-coding RNAs, have been shown to regulate the development and function of the hippocampus, and the alteration of epigenetic regulation may play important roles in the development of neurocognitive and neurodegenerative diseases. This review summarizes the epigenetic modifications of various cell types and processes within the hippocampus and their resulting effects on cognition, memory and overall hippocampal function. In addition, the effects of exposure to radiation that may induce a myriad of epigenetic changes in the hippocampus are reviewed. By assessing and evaluating the current literature, we hope to prompt a more thorough understanding of the molecular mechanisms that underlie radiation-induced epigenetic changes, an area which can be further explored.
Collapse
|
37
|
Quarta A, Berneman Z, Ponsaerts P. Functional consequences of a close encounter between microglia and brain-infiltrating monocytes during CNS pathology and repair. J Leukoc Biol 2020; 110:89-106. [PMID: 33155726 DOI: 10.1002/jlb.3ru0820-536r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is recognized as an important factor contributing to the development and progression of several central nervous system (CNS) disorders. Upon CNS trauma or disease, parenchymal microglia highly proliferate and accumulate in and around the lesion site. In addition, blood-derived monocytes can infiltrate the inflamed CNS in response to cellular damage and/or a compromised blood-brain barrier. Both microglia and infiltrating monocytes are characterized by multiple functional states and can either display highly proinflammatory properties or promote resolution of inflammation and tissue regeneration. Despite sharing some basic immunologic functions, microglia and monocytes display many distinctive features, which ultimately define their contribution to neuropathology. Understanding how the innate immune system participates to brain disease is imperative to identify novel treatment options for CNS inflammatory disorders. In this context, existing and newly developed in vitro platforms for disease modeling are fundamental tools to investigate and modulate microglia and monocyte immune functions within a specific neuropathologic context. In this review, we first briefly summarize the current knowledge on microglia and monocyte ontogenesis, as well as their complex and interconnected contributions to the development of various CNS pathologies. Following the well-recognized concept that both microglia and monocytes can either exert neuroprotective functions or exacerbate tissue damage, we provide a comprehensive overview of cellular models currently available for in vitro study of neuroinflammatory responses. In this context, we highlight how simplified single-cell models may not always correctly recapitulate in vivo biology, hence future research should move toward novel models with higher and multicellular complexity.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
38
|
Abstract
The innate immune system in the central nervous system (CNS) is mainly represented by specialized tissue-resident macrophages, called microglia. In the past years, various species-, host- and tissue-specific as well as environmental factors were recognized that essentially affect microglial properties and functions in the healthy and diseased brain. Host microbiota are mostly residing in the gut and contribute to microglial activation states, for example, via short-chain fatty acids (SCFAs) or aryl hydrocarbon receptor (AhR) ligands. Thereby, the gut microorganisms are deemed to influence numerous CNS diseases mediated by microglia. In this review, we summarize recent findings of the interaction between the host microbiota and the CNS in health and disease, where we specifically highlight the resident gut microbiota as a crucial environmental factor for microglial function as what we coin "the microbiota-microglia axis."
Collapse
Affiliation(s)
- Omar Mossad
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Daniel Erny
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
| |
Collapse
|
39
|
Béchade C, D'Andrea I, Etienne F, Verdonk F, Moutkine I, Banas SM, Kolodziejczak M, Diaz SL, Parkhurst CN, Gan WB, Maroteaux L, Roumier A. The serotonin 2B receptor is required in neonatal microglia to limit neuroinflammation and sickness behavior in adulthood. Glia 2020; 69:638-654. [PMID: 33095507 DOI: 10.1002/glia.23918] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/11/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022]
Abstract
Severe peripheral infections induce an adaptive sickness behavior and an innate immune reaction in various organs including the brain. On the long term, persistent alteration of microglia, the brain innate immune cells, is associated with an increased risk of psychiatric disorders. It is thus critical to identify genes and mechanisms controlling the intensity and duration of the neuroinflammation induced by peripheral immune challenges. We tested the hypothesis that the 5-HT2B receptor, the main serotonin receptor expressed by microglia, might represent a valuable candidate. First, we observed that Htr2b-/- mice, knock-out for the 5-HT2B receptor gene, developed, when exposed to a peripheral lipopolysaccharide (LPS) challenge, a stronger weight loss compared to wild-type mice; in addition, comparison of inflammatory markers in brain, 4 and 24 hr after LPS injection, showed that Htr2b deficiency leads to a prolonged neuroinflammation. Second, to assess the specific contribution of the microglial 5-HT2B receptor, we investigated the response to LPS of conditional knock-out mice invalidated for Htr2b in microglia only. We found that deletion of Htr2b in microglia since birth is sufficient to cause enhanced weight loss and increased neuroinflammatory response upon LPS injection at adult stage. In contrast, mice deleted for microglial Htr2b in adulthood responded normally to LPS, revealing a neonatal developmental effect. These results highlight the role of microglia in the response to a peripheral immune challenge and suggest the existence of a developmental, neonatal period, during which instruction of microglia through 5-HT2B receptors is necessary to prevent microglia overreactivity in adulthood.
Collapse
Affiliation(s)
- Catherine Béchade
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivana D'Andrea
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Fanny Etienne
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Franck Verdonk
- Experimental Neuropathology, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - Imane Moutkine
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Sophie M Banas
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marta Kolodziejczak
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Silvina L Diaz
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Christopher N Parkhurst
- Molecular Neurobiology Program, The Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, USA
| | - Wenbiao B Gan
- Molecular Neurobiology Program, The Kimmel Center for Biology and Medicine at the Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, USA
| | - Luc Maroteaux
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Anne Roumier
- INSERM UMR-S 1270, Paris, France.,Sorbonne Université, Faculté des Sciences et Ingénierie, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
40
|
Nelson MC, O'Connell RM. MicroRNAs: At the Interface of Metabolic Pathways and Inflammatory Responses by Macrophages. Front Immunol 2020; 11:1797. [PMID: 32922393 PMCID: PMC7456828 DOI: 10.3389/fimmu.2020.01797] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages are key cells of the innate immune system with functional roles in both homeostatic maintenance of self-tissues and inflammatory responses to external stimuli, including infectious agents. Recent advances in metabolic research have revealed that macrophage functions rely upon coordinated metabolic programs to regulate gene expression, inflammation, and other important cellular processes. Polarized macrophages adjust their use of nutrients such as glucose and amino acids to meet their changing metabolic needs, and this in turn supports the functions of the activated macrophage. Metabolic and inflammatory processes have been widely studied, and a crucial role for their regulation at the post-transcriptional level by microRNAs (miRNAs) has been identified. miRNAs govern many facets of macrophage biology, including direct targeting of metabolic regulators and inflammatory pathways. This review will integrate emerging data that support an interplay between miRNAs and metabolism during macrophage inflammatory responses, highlighting critical miRNAs and miRNA families. Additionally, we will address the implications of these networks for human disease and discuss emerging areas of research in this field.
Collapse
Affiliation(s)
- Morgan C Nelson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
41
|
Brain Dicer1 Is Down-Regulated in a Mouse Model of Alzheimer's Disease Via Aβ42-Induced Repression of Nuclear Factor Erythroid 2-Related Factor 2. Mol Neurobiol 2020; 57:4417-4437. [PMID: 32737764 DOI: 10.1007/s12035-020-02036-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022]
Abstract
Dicer1 is a microRNA-processing enzyme which plays critical roles in neuronal survival and neuritogenesis. Dicer1 deletion induces neurodegeneration or degeneration in retinal pigment epithelium, which is associated with oxidative stress. Oxidative stress is thought to be central in the pathogenesis of Alzheimer's disease (AD). Therefore, we hypothesize that Dicer1 may play roles in AD. Using immunoblotting and quantitative real-time PCR, Dicer1 protein and mRNA were reduced in the hippocampi of the AD mouse model APPswe/PSEN1dE9 compared with littermate controls. SiRNA-mediated Dicer1 knockdown induced oxidative stress and apoptosis and reduced mitochondrial membrane potential in cultured neurons. Chronic Aβ42 exposure decreased Dicer1 and nuclear factor erythroid 2-related factor 2 (Nrf2) which were reversed by N-acetyl-cystein. Nrf2 overexpression increased Dicer1 mRNA and protein and reverted the Aβ42-induced Dicer1 reduction. We further cloned Dicer1 promoter variants harboring the Nrf2-binding site, the antioxidant response elements (ARE), into a luciferase reporter and found that simultaneous transfection of Nrf2-expressing plasmid increased luciferase expression from these promoter constructs. ChIP assays indicated that Nrf2 directly interacted with the ARE motifs in the Dicer1 promoter. Furthermore, Dicer1 overexpression in cultured neurons reverted Aβ42-induced neurite deficits. Notably, injection of Dicer1-expressing adenovirus into the hippocampus of the mice significantly improved spatial learning. Altogether, we found novel roles of Dicer1 in AD and a novel regulatory pathway for Dicer1. These results suggest that Dicer1 is a target in AD therapy, especially at the early stage of this disorder. In this study, we found that Dicer1 was reduced in the brain of AD mice which is the first report to examine Dicer1 in AD. We further found (i) that Aβ42 exposure decreased Dicer1 via attenuating Nrf2-ARE signaling and (ii) injection of Dicer1-expressing adenovirus into the hippocampus of the AD mice significantly improved spatial learning. Altogether, we found novel roles of Dicer1 in AD and a novel regulatory pathway for Dicer1. This study may open new avenues for investigating potential pathognomonics and pathogenesis in AD.
Collapse
|
42
|
Althammer F, Ferreira-Neto HC, Rubaharan M, Roy RK, Patel AA, Murphy A, Cox DN, Stern JE. Three-dimensional morphometric analysis reveals time-dependent structural changes in microglia and astrocytes in the central amygdala and hypothalamic paraventricular nucleus of heart failure rats. J Neuroinflammation 2020; 17:221. [PMID: 32703230 PMCID: PMC7379770 DOI: 10.1186/s12974-020-01892-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiovascular diseases, including heart failure, are the most common cause of death globally. Recent studies support a high degree of comorbidity between heart failure and cognitive and mood disorders resulting in memory loss, depression, and anxiety. While neuroinflammation in the hypothalamic paraventricular nucleus contributes to autonomic and cardiovascular dysregulation in heart failure, mechanisms underlying cognitive and mood disorders in this disease remain elusive. The goal of this study was to quantitatively assess markers of neuroinflammation (glial morphology, cytokines, and A1 astrocyte markers) in the central amygdala, a critical forebrain region involved in emotion and cognition, and to determine its time course and correlation to disease severity during the progression of heart failure. METHODS We developed and implemented a comprehensive microglial/astrocyte profiler for precise three-dimensional morphometric analysis of individual microglia and astrocytes in specific brain nuclei at different time points during the progression of heart failure. To this end, we used a well-established ischemic heart failure rat model. Morphometric studies were complemented with quantification of various pro-inflammatory cytokines and A1/A2 astrocyte markers via qPCR. RESULTS We report structural remodeling of central amygdala microglia and astrocytes during heart failure that affected cell volume, surface area, filament length, and glial branches, resulting overall in somatic swelling and deramification, indicative of a change in glial state. These changes occurred in a time-dependent manner, correlated with the severity of heart failure, and were delayed compared to changes in the hypothalamic paraventricular nucleus. Morphometric changes correlated with elevated mRNA levels of pro-inflammatory cytokines and markers of reactive A1-type astrocytes in the paraventricular nucleus and central amygdala during heart failure. CONCLUSION We provide evidence that in addition to the previously described hypothalamic neuroinflammation implicated in sympathohumoral activation during heart failure, microglia, and astrocytes within the central amygdala also undergo structural remodeling indicative of glial shifts towards pro-inflammatory phenotypes. Thus, our studies suggest that neuroinflammation in the amygdala stands as a novel pathophysiological mechanism and potential therapeutic target that could be associated with emotional and cognitive deficits commonly observed at later stages during the course of heart failure.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | | | | | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Anne Murphy
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA.
| |
Collapse
|
43
|
Prinz M, Jung S, Priller J. Microglia Biology: One Century of Evolving Concepts. Cell 2020; 179:292-311. [PMID: 31585077 DOI: 10.1016/j.cell.2019.08.053] [Citation(s) in RCA: 918] [Impact Index Per Article: 183.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 01/05/2023]
Abstract
Microglia were first recognized as a distinct cell population in the CNS one century ago. For a long time, they were primarily considered to be phagocytes responsible for removing debris during CNS development and disease. More recently, advances in imaging and genetics and the advent of single-cell technologies provided new insights into the much more complex and fascinating biology of microglia. The ontogeny of microglia was identified, and their functions in health and disease were better defined. Although many questions about microglia and their roles in human diseases remain unanswered, the prospect of targeting microglia for the treatment of neurological and psychiatric disorders is tantalizing.
Collapse
Affiliation(s)
- Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany; DZNE and BIH, Berlin, Germany; University of Edinburgh and UK DRI, Edinburgh, UK.
| |
Collapse
|
44
|
Guo CH, Cao T, Zheng LT, Waddington JL, Zhen XC. Development and characterization of an inducible Dicer conditional knockout mouse model of Parkinson's disease: validation of the antiparkinsonian effects of a sigma-1 receptor agonist and dihydromyricetin. Acta Pharmacol Sin 2020; 41:499-507. [PMID: 32112040 PMCID: PMC7468551 DOI: 10.1038/s41401-020-0379-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by motor impairment and progressive loss of dopamine (DA) neurons. At present, the acute application of neurotoxic drugs such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA) are commonly used to simulate the pathology of PD; however, it is difficult to induce the progressive pathogenesis of PD with these models. In this study, we employed DAT promoter-mediated Cre transgenic mice to establish tamoxifen-inducible Dicer conditional knockout (cKO) mice in an effort to mimic the progressive loss of DA neurons and the development of PD-like behavioral phenotypes. The results showed that Dicer cKO mice exhibited progressive loss of DA neurons in the substantia nigra (SN) following tamoxifen administration. Significant DA loss was observed 6 weeks after tamoxifen administration; accordingly, progressive motor function impairment was also observed. We also found that a significant neuroinflammatory response, as evidenced by microglial proliferation, another hallmark of PD pathogenesis, accompanied the loss of DA neurons. The acute application of levo-DOPA (L-DOPA) relieved the PD-like motor impairments in Dicer cKO mice to exert its antiparkinsonian action, indicating that the model can be used to evaluate the antiparkinsonian efficacy of PD drugs. To further elucidate the potential application of this novel PD animal model for PD drug development, we employed the powerful neuroprotective agent dihydromyricetin (DHM) (10 mg/kg) and the selective sigma-1 receptor agonist PRE-084 (1 mg/kg), both of which were previously shown to produce antiparkinsonian effects. The results indicated that the chronic administration of either DHM or PRE-084 attenuated the Dicer cKO-induced loss of DA neurons and motor impairments, although the two drugs acted through different mechanisms. These data indicate that the Dicer cKO mouse model may be a useful model for investigating the pathological development of PD and intervention-mediated changes. In conclusion, this transgenic mouse model appears to simulate the progressive pathogenesis of PD and may be a potentially useful model for PD drug discovery.
Collapse
Affiliation(s)
- Chen-Hong Guo
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ting Cao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Long-Tai Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - John L Waddington
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
- School of Pharmacy and Biomolecular Sciences (PBS), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
45
|
Murgoci AN, Duhamel M, Raffo-Romero A, Mallah K, Aboulouard S, Lefebvre C, Kobeissy F, Fournier I, Zilkova M, Maderova D, Cizek M, Cizkova D, Salzet M. Location of neonatal microglia drives small extracellular vesicles content and biological functions in vitro. J Extracell Vesicles 2020; 9:1727637. [PMID: 32158520 PMCID: PMC7049881 DOI: 10.1080/20013078.2020.1727637] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Combining proteomics and systems biology approaches, we demonstrate that neonatal microglial cells derived from two different CNS locations, cortex and spinal cord, and cultured in vitro displayed different phenotypes upon different physiological or pathological conditions. These cells also exhibited greater variability in terms of cellular and small extracellular vesicles (sEVs) protein content and levels. Bioinformatic data analysis showed that cortical microglia exerted anti-inflammatory and neurogenesis/tumorigenesis properties, while the spinal cord microglia were more inflammatory. Interestingly, while both sEVs microglia sources enhanced growth of DRGs processes, only the spinal cord-derived sEVs microglia under LPS stimulation significantly attenuated glioma proliferation. These results were confirmed using the neurite outgrowth assay on DRGs cells and glioma proliferation analysis in 3D spheroid cultures. Results from these in vitro assays suggest that the microglia localized at different CNS regions can ensure different biological functions. Together, this study indicates that neonatal microglia locations regulate their physiological and pathological functional fates and could affect the high prevalence of brain vs spinal cord gliomas in adults.
Collapse
Affiliation(s)
- Adriana-Natalia Murgoci
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Marie Duhamel
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Antonella Raffo-Romero
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Khalil Mallah
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Soulaimane Aboulouard
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Christophe Lefebvre
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Firas Kobeissy
- Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Isabelle Fournier
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Monika Zilkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Denisa Maderova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Milan Cizek
- Department of Epizootiology and Parasitology, University of Veterinary Medicine and Pharmacy in Košice, KošiceSlovakia
| | - Dasa Cizkova
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Michel Salzet
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| |
Collapse
|
46
|
Gutbrod MJ, Martienssen RA. Conserved chromosomal functions of RNA interference. Nat Rev Genet 2020; 21:311-331. [PMID: 32051563 DOI: 10.1038/s41576-019-0203-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2019] [Indexed: 12/21/2022]
Abstract
RNA interference (RNAi), a cellular process through which small RNAs target and regulate complementary RNA transcripts, has well-characterized roles in post-transcriptional gene regulation and transposon repression. Recent studies have revealed additional conserved roles for RNAi proteins, such as Argonaute and Dicer, in chromosome function. By guiding chromatin modification, RNAi components promote chromosome segregation during both mitosis and meiosis and regulate chromosomal and genomic dosage response. Small RNAs and the RNAi machinery also participate in the resolution of DNA damage. Interestingly, many of these lesser-studied functions seem to be more strongly conserved across eukaryotes than are well-characterized functions such as the processing of microRNAs. These findings have implications for the evolution of RNAi since the last eukaryotic common ancestor, and they provide a more complete view of the functions of RNAi.
Collapse
Affiliation(s)
- Michael J Gutbrod
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Robert A Martienssen
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA. .,Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
47
|
Brioschi S, Zhou Y, Colonna M. Brain Parenchymal and Extraparenchymal Macrophages in Development, Homeostasis, and Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:294-305. [PMID: 31907272 PMCID: PMC7034672 DOI: 10.4049/jimmunol.1900821] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Microglia are parenchymal macrophages of the CNS; as professional phagocytes they are important for maintenance of the brain's physiology. These cells are generated through primitive hematopoiesis in the yolk sac and migrate into the brain rudiment after establishment of embryonic circulation. Thereafter, microglia develop in a stepwise fashion, reaching complete maturity after birth. In the CNS, microglia self-renew without input from blood monocytes. Recent RNA-sequencing studies have defined a molecular signature for microglia under homeostasis. However, during disease, microglia undergo remarkable phenotypic changes, which reflect the acquisition of specialized functions tailored to the pathological context. In addition to microglia, the brain-border regions host populations of extraparenchymal macrophages with disparate origins and phenotypes that have recently been delineated. In this review we outline recent findings that provide a deeper understanding of both parenchymal microglia and extraparenchymal brain macrophages in homeostasis and during disease.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
48
|
Tang FL, Zhao L, Zhao Y, Sun D, Zhu XJ, Mei L, Xiong WC. Coupling of terminal differentiation deficit with neurodegenerative pathology in Vps35-deficient pyramidal neurons. Cell Death Differ 2020; 27:2099-2116. [PMID: 31907392 PMCID: PMC7308361 DOI: 10.1038/s41418-019-0487-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Vps35 (vacuolar protein sorting 35) is a key component of retromer that regulates transmembrane protein trafficking. Dysfunctional Vps35 is a risk factor for neurodegenerative diseases, including Parkinson’s and Alzheimer’s diseases. Vps35 is highly expressed in developing pyramidal neurons, and its physiological role in developing neurons remains to be explored. Here, we provide evidence that Vps35 in embryonic neurons is necessary for axonal and dendritic terminal differentiation. Loss of Vps35 in embryonic neurons results in not only terminal differentiation deficits, but also neurodegenerative pathology, such as cortical brain atrophy and reactive glial responses. The atrophy of neocortex appears to be in association with increases in neuronal death, autophagosome proteins (LC3-II and P62), and neurodegeneration associated proteins (TDP43 and ubiquitin-conjugated proteins). Further studies reveal an increase of retromer cargo protein, sortilin1 (Sort1), in lysosomes of Vps35-KO neurons, and lysosomal dysfunction. Suppression of Sort1 diminishes Vps35-KO-induced dendritic defects. Expression of lysosomal Sort1 recapitulates Vps35-KO-induced phenotypes. Together, these results demonstrate embryonic neuronal Vps35’s function in terminal axonal and dendritic differentiation, reveal an association of terminal differentiation deficit with neurodegenerative pathology, and uncover an important lysosomal contribution to both events.
Collapse
Affiliation(s)
- Fu-Lei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia
| | - Lu Zhao
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia.,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.,Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Yang Zhao
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.,Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Dong Sun
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia.,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Xiao-Juan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Lin Mei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia.,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia. .,Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
49
|
Kodama L, Guzman E, Etchegaray JI, Li Y, Sayed FA, Zhou L, Zhou Y, Zhan L, Le D, Udeochu JC, Clelland CD, Cheng Z, Yu G, Li Q, Kosik KS, Gan L. Microglial microRNAs mediate sex-specific responses to tau pathology. Nat Neurosci 2019; 23:167-171. [PMID: 31873194 PMCID: PMC7394069 DOI: 10.1038/s41593-019-0560-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/20/2019] [Indexed: 12/16/2022]
Abstract
Sex is a key modifier of neurological disease outcomes. Microglia are implicated in neurological diseases and modulated by miRNAs, but it is unknown whether microglial miRNAs have sex-specific influences on disease. We show that microglial miRNA expression differs in males and females and that loss of miRNAs leads to sex-specific changes in the microglial transcriptome and tau pathology. These findings suggest microglial miRNAs influence tau pathogenesis in a sex-specific manner.
Collapse
Affiliation(s)
- Lay Kodama
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.,Gladstone Institute of Neurological Disease, San Francisco, CA, USA.,Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, NY, New York, USA.,Medical Scientist Training Program and Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Elmer Guzman
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA.,Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jon I Etchegaray
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Faten A Sayed
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.,Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Lu Zhou
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Lihong Zhan
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - David Le
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Joe C Udeochu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, NY, New York, USA
| | - Claire D Clelland
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.,Department of Neurobiology, University of California, San Francisco, San Francisco, CA, USA
| | - Zuolin Cheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Guoqiang Yu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, USA
| | - Qingyun Li
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kenneth S Kosik
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA.,Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Li Gan
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA. .,Gladstone Institute of Neurological Disease, San Francisco, CA, USA. .,Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, NY, New York, USA.
| |
Collapse
|
50
|
When encephalitogenic T cells collaborate with microglia in multiple sclerosis. Nat Rev Neurol 2019; 15:704-717. [PMID: 31527807 DOI: 10.1038/s41582-019-0253-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2019] [Indexed: 01/07/2023]
Abstract
Immune cells mediate critical inflammatory and neurodegenerative processes in the CNS in individuals with multiple sclerosis (MS). In MS, activated microglia, border-associated macrophages and monocyte-derived macrophages in the CNS can encounter T cells that have infiltrated the brain parenchyma from the circulation. Although microglia and T cells both contribute to normal CNS development and homeostasis, evidence suggests that the meeting of activated microglia and macrophages with encephalitogenic T cells exacerbates their capacity to inflict injury. This crosstalk involves many cell-surface molecules, cytokines and neurotoxic factors. In this Review, we summarize the mechanisms and consequences of T cell-microglia interactions as identified with in vitro experiments and animal models, and discuss the challenges that arise when translating this preclinical knowledge to MS in humans. We also consider therapeutic approaches to MS of which the mechanisms involve prevention or modulation of T cell and microglia responses and their interactions.
Collapse
|