1
|
Xue S, Yi P, Mao Y, Zhan Z, Cai Y, Song Z, Wang K, Yang K, Song Y, Wang X, Long H. Nucleus accumbens shell electrical lesion attenuates seizures and gliosis in chronic temporal lobe epilepsy rats. Epileptic Disord 2024. [PMID: 39570088 DOI: 10.1002/epd2.20316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE Temporal lobe epilepsy (TLE) is the most prevalent form of epilepsy. Prior research has indicated the involvement of the nucleus accumbens shell (NAcSh) in the process of epileptogenesis, thereby implying its potential as a therapeutic target for TLE. In the present study, we investigated the antiepileptic effect of the NAcSh electrical lesion. METHODS Chronic TLE was induced by stereotactic injection of kainic acid (KA) into the hippocampus 3 weeks after KA administration, and NAcSh electrical lesions were performed. Seizures in rats were monitored by video electroencephalogram (EEG) 1 week following the NAcSh electrical lesion. Besides, the spatial memory function assessment in rats was conducted using the Morris water maze (MWM) test in the final week of the experiment. Later, hippocampal glial cell activation and neuron loss in rats were evaluated through immunohistochemistry. RESULTS TLE rats subjected to NAcSh electrical lesion exhibited a significant reduction in the frequency of seizures compared to untreated TLE rats. Furthermore, NAcSh electrical lesion led to less activation of hippocampal glial cells and fewer neuronal loss in TLE rats. It is worth noting that the NAcSh electrical lesion did not cause additional memory impairment. SIGNIFICANCE In the present study, the NAcSh electrical lesion exhibited a definitive therapeutic effect on the chronic TLE rat model, potentially due to decreased hippocampal TLE-induced activation of glial cells and neuron loss. In conclusion, our results indicated that the NAcSh is a promising therapeutic target for TLE and possesses high potential for clinical application.
Collapse
Affiliation(s)
- Shuaishuai Xue
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Peiyao Yi
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yangqi Mao
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhengming Zhan
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yonghua Cai
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zibin Song
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Kewan Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Kaijun Yang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Ye Song
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Xingqin Wang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Hao Long
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Smail MA, Lenz KM. Developmental functions of microglia: Impact of psychosocial and physiological early life stress. Neuropharmacology 2024; 258:110084. [PMID: 39025401 DOI: 10.1016/j.neuropharm.2024.110084] [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: 02/15/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Microglia play numerous important roles in brain development. From early embryonic stages through adolescence, these immune cells influence neuronal genesis and maturation, guide connectivity, and shape brain circuits. They also interact with other glial cells and structures, influencing the brain's supportive microenvironment. While this central role makes microglia essential, it means that early life perturbations to microglia can have widespread effects on brain development, potentially resulting in long-lasting behavioral impairments. Here, we will focus on the effects of early life psychosocial versus physiological stressors in rodent models. Psychosocial stress refers to perceived threats that lead to stress axes activation, including prenatal stress, or chronic postnatal stress, including maternal separation and resource scarcity. Physiological stress refers to physical threats, including maternal immune activation, postnatal infection, and traumatic brain injury. Differing sources of early life stress have varied impacts on microglia, and these effects are moderated by factors such as developmental age, brain region, and sex. Overall, these stressors appear to either 1) upregulate basal microglia numbers and activity throughout the lifespan, while possibly blunting their responsivity to subsequent stressors, or 2) shift the developmental curve of microglia, resulting in differential timing and function, impacting the critical periods they govern. Either could contribute to behavioral dysfunctions that occur after the resolution of early life stress. Exploring how different stressors impact microglia, as well as how multiple stressors interact to alter microglia's developmental functions, could deepen our understanding of how early life stress changes the brain's developmental trajectory. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Psychology, Ohio State University, Columbus, OH, USA.
| | - Kathryn M Lenz
- Department of Psychology, Ohio State University, Columbus, OH, USA; Department of Neuroscience, Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, Ohio State University, Columbus, OH, USA
| |
Collapse
|
3
|
Zheng C, Xiao X, Zhao W, Yang Z, Guo S. Functional brain network controllability dysfunction in Alzheimer's disease and its relationship with cognition and gene expression profiling. J Neural Eng 2024; 21:026018. [PMID: 38502960 DOI: 10.1088/1741-2552/ad357e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/19/2024] [Indexed: 03/21/2024]
Abstract
Objective. In recent studies, network control theory has been applied to clarify transitions between brain states, emphasizing the significance of assessing the controllability of brain networks in facilitating transitions from one state to another. Despite these advancements, the potential alterations in functional network controllability associated with Alzheimer's disease (AD), along with the underlying genetic mechanisms responsible for these alterations, remain unclear.Approach. We conducted a comparative analysis of functional network controllability measures between patients with AD (n= 64) and matched normal controls (NCs,n= 64). We investigated the association between altered controllability measures and cognitive function in AD. Additionally, we conducted correlation analyses in conjunction with the Allen Human Brain Atlas to identify genes whose expression was correlated with changes in functional network controllability in AD, followed by a set of analyses on the functional features of the identified genes.Main results. In comparison to NCs, patients with AD exhibited a reduction in average controllability, predominantly within the default mode network (DMN) (63% of parcellations), and an increase in average controllability within the limbic (LIM) network (33% of parcellations). Conversely, AD patients displayed a decrease in modal controllability within the LIM network (27% of parcellations) and an increase in modal controllability within the DMN (80% of parcellations). In AD patients, a significant positive correlation was found between the average controllability of the salience network and the mini-mental state examination scores. The changes in controllability measures exhibited spatial correlation with transcriptome profiles. The significant genes identified exhibited enrichment in neurobiologically relevant pathways and demonstrated preferential expression in various tissues, cell types, and developmental periods.Significance. Our findings have the potential to offer new insights into the genetic mechanisms underlying alterations in the controllability of functional networks in AD. Additionally, these results offered perspectives for a deeper understanding of the pathogenesis and the development of therapeutic strategies for AD.
Collapse
Affiliation(s)
- Chuchu Zheng
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Xiaoxia Xiao
- School of Informatics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China
| | - Wei Zhao
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Zeyu Yang
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Shuixia Guo
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| |
Collapse
|
4
|
Rezaie P, Hanisch UK. History of Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:15-37. [PMID: 39207684 DOI: 10.1007/978-3-031-55529-9_2] [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
The term 'microglia' was first introduced into the scientific literature a century ago. The various eras of microglial research have been defined not only by the number of reports subsequently generated but, more critically, also by the concepts that have shaped our present-day views and understanding of microglia. Key methods, technologies, and models, as well as seminal discoveries made possible through their deployment have enabled breakthroughs, and now pave the way for lines of investigation that could not have been anticipated even a decade ago. Advances in our understanding of the microglial origin, forms, and functions have relied fundamentally on parallel developments in immunology. As the 'neuro-immune' cells of the brain, microglia are now under the spotlight in various disciplines. This chapter surveys the gradual processes and precipitous events that helped form ideas concerning the developmental origin of microglia and their roles in health and disease. It first covers the dawning phase during which the early pioneers of microglial research discovered cellular entities and already assigned functions to them. Following a recess period, the 1960s brought about a renaissance of active interest, with the development of tools and models-and fundamental notions on microglial contributions to central nervous system (CNS) pathologies. These seminal efforts laid the foundation for the awakening of a sweeping research era beginning in the 1980s and spurred on by a blast of immunological discoveries. Finally, this chapter stresses the advancements in molecular, genetic, and imaging approaches to the study of microglia with the turn of the millennium, enabling insights into virtually all facets of microglial physiology. Moving forward, it is clear that the future holds substantial promise for further discoveries. The next epoch in the history of microglial research has just begun.
Collapse
Affiliation(s)
- Payam Rezaie
- School of Life, Health & Chemical Sciences, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, UK.
| | | |
Collapse
|
5
|
Pinho AG, Monteiro A, Fernandes S, de Sousa N, Salgado AJ, Silva NA, Monteiro S. The Central Nervous System Source Modulates Microglia Function and Morphology In Vitro. Int J Mol Sci 2023; 24:ijms24097685. [PMID: 37175391 PMCID: PMC10177862 DOI: 10.3390/ijms24097685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The regional heterogeneity of microglia was first described a century ago by Pio del Rio Hortega. Currently, new information on microglia heterogeneity throughout central nervous system (CNS) regions is being revealed by high-throughput techniques. It remains unclear whether these spatial specificities translate into different microglial behaviors in vitro. We cultured microglia isolated from the cortex and spinal cord and analyzed the effect of the CNS spatial source on behavior in vitro by applying the same experimental protocol and culture conditions. We analyzed the microglial cell numbers, function, and morphology and found a distinctive in vitro phenotype. We found that microglia were present in higher numbers in the spinal-cord-derived glial cultures, presenting different expressions of inflammatory genes and a lower phagocytosis rate under basal conditions or after activation with LPS and IFN-γ. Morphologically, the cortical microglial cells were more complex and presented longer ramifications, which were also observed in vivo in CX3CR1+/GFP transgenic reporter mice. Collectively, our data demonstrated that microglial behavior in vitro is defined according to specific spatial characteristics acquired by the tissue. Thus, our study highlights the importance of microglia as a source of CNS for in vitro studies.
Collapse
Affiliation(s)
- Andreia G Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Andreia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Sara Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Nídia de Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
6
|
Hu X, Geng P, Zhao X, Wang Q, Liu C, Guo C, Dong W, Jin X. The NG2-glia is a potential target to maintain the integrity of neurovascular unit after acute ischemic stroke. Neurobiol Dis 2023; 180:106076. [PMID: 36921779 DOI: 10.1016/j.nbd.2023.106076] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
The neurovascular unit (NVU) plays a critical role in health and disease. In the current review, we discuss the critical role of a class of neural/glial antigen 2 (NG2)-expressing glial cells (NG2-glia) in regulating NVU after acute ischemic stroke (AIS). We first introduce the role of NG2-glia in the formation of NVU during development as well as aging-induced damage to NVU and accompanying NG2-glia change. We then discuss the reciprocal interactions between NG2-glia and the other component cells of NVU, emphasizing the factors that could influence NG2-glia. Damage to the NVU integrity is the pathological basis of edema and hemorrhagic transformation, the most dreaded complication after AIS. The role of NG2-glia in AIS-induced NVU damage and the effect of NG2-glia transplantation on AIS-induced NVU damage are summarized. We next discuss the role of NG2-glia and the effect of NG2-glia transplantation in oligodendrogenesis and white matter repair as well as angiogenesis which is associated with the outcome of the patients after AIS. Finally, we review the current strategies to promote NG2-glia proliferation and differentiation and propose to use the dental pulp stem cells (DPSC)-derived exosome as a promising strategy to reduce AIS-induced injury and promote repair through maintaining the integrity of NVU by regulating endogenous NG2-glia proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaoyan Hu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Xiaoyun Zhao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Changqing Liu
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Wen Dong
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
7
|
Caruso G, Privitera A, Saab MW, Musso N, Maugeri S, Fidilio A, Privitera AP, Pittalà A, Jolivet RB, Lanzanò L, Lazzarino G, Caraci F, Amorini AM. Characterization of Carnosine Effect on Human Microglial Cells under Basal Conditions. Biomedicines 2023; 11:biomedicines11020474. [PMID: 36831010 PMCID: PMC9953171 DOI: 10.3390/biomedicines11020474] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
The activity of microglia is fundamental for the regulation of numerous physiological processes including brain development, synaptic plasticity, and neurogenesis, and its deviation from homeostasis can lead to pathological conditions, including numerous neurodegenerative disorders. Carnosine is a naturally occurring molecule with well-characterized antioxidant and anti-inflammatory activities, able to modulate the response and polarization of immune cells and ameliorate their cellular energy metabolism. The better understanding of microglia characteristics under basal physiological conditions, as well as the possible modulation of the mechanisms related to its response to environmental challenges and/or pro-inflammatory/pro-oxidant stimuli, are of utmost importance for the development of therapeutic strategies. In the present study, we assessed the activity of carnosine on human HMC3 microglial cells, first investigating the effects of increasing concentrations of carnosine on cell viability. When used at a concentration of 20 mM, carnosine led to a decrease of cell viability, paralleled by gene expression increase and decrease, respectively, of interleukin 6 and heme oxygenase 1. When using the maximal non-toxic concentration (10 mM), carnosine decreased nitric oxide bioavailability, with no changes in the intracellular levels of superoxide ion. The characterization of energy metabolism of HMC3 microglial cells under basal conditions, never reported before, demonstrated that it is mainly based on mitochondrial oxidative metabolism, paralleled by a high rate of biosynthetic reactions. The exposure of HMC3 cells to carnosine seems to ameliorate microglia energy state, as indicated by the increase in the adenosine triphosphate/adenosine diphosphate (ATP/ADP) ratio and energy charge potential. The improvement of cell energy metabolism mediated by 10 mM carnosine could represent a useful protective weapon in the case of human microglia undergoing stressing conditions.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Correspondence: ; Tel.: +39-0957385036
| | - Anna Privitera
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Miriam Wissam Saab
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Salvatore Maugeri
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Annamaria Fidilio
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | | | - Alessandra Pittalà
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Renaud Blaise Jolivet
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Luca Lanzanò
- Department of Physics and Astronomy “Ettore Majorana”, University of Catania, 95123 Catania, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Angela Maria Amorini
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| |
Collapse
|
8
|
Abd El-Wahab AHF, Mohamed HM. Synthesis and DFT Study of 7-Bromophenylnaphthopyran Moieties. ASIAN JOURNAL OF CHEMISTRY 2023; 35:1819-1826. [DOI: 10.14233/ajchem.2023.28032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
A one-pot, three-component reaction of 6-bromo-2-naphthol (1), p-chlorobenzaldehyde (2) and
malononitrile or ethyl cyanoacetate (3) in ethanol/piperidine under reflux was performed to afford
4H-naphtho[2,1-b]pyrano-3-carbonitrile (4a) and ethyl 4H-naphtho[2,1-b]pyrano-3-carboxylate (4b)
derivatives, respectively. The structure of these compounds was determined using IR, 1H NMR, 13C
NMR, mass spectroscopy and UV-Vis spectra. The molecular geometry of compounds 4a and 4b was
determined at the B3LYP/631+G(d) level. The geometric optimization was performed on two tautomers
and two conformers. Tautomers were separated by about 7.942 kcal/mol, while rotational conformers
were separated by just 0.511 kcal/mol. The global electrophilicity, hardness, softness and local
condensed Fukui functions were calculated and considered as molecular reactivity descriptors, moreover
the frontier molecular orbitals (HOMO and LUMO) were also calculated.
Collapse
Affiliation(s)
| | - Hany Mostafa Mohamed
- Chemistry Department, Faculty of Science, Jazan University, 2097, Jazan, Saudi Arabia
| |
Collapse
|
9
|
Wong JHC, Ma JYW, Jobling AI, Brandli A, Greferath U, Fletcher EL, Vessey KA. Exploring the pathogenesis of age-related macular degeneration: A review of the interplay between retinal pigment epithelium dysfunction and the innate immune system. Front Neurosci 2022; 16:1009599. [PMID: 36408381 PMCID: PMC9670140 DOI: 10.3389/fnins.2022.1009599] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/12/2022] [Indexed: 07/30/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of irreversible vision loss in the older population. Classical hallmarks of early and intermediate AMD are accumulation of drusen, a waste deposit formed under the retina, and pigmentary abnormalities in the retinal pigment epithelium (RPE). When the disease progresses into late AMD, vision is affected due to death of the RPE and the light-sensitive photoreceptors. The RPE is essential to the health of the retina as it forms the outer blood retinal barrier, which establishes ocular immune regulation, and provides support for the photoreceptors. Due to its unique anatomical position, the RPE can communicate with the retinal environment and the systemic immune environment. In AMD, RPE dysfunction and the accumulation of drusen drive the infiltration of retinal and systemic innate immune cells into the outer retina. While recruited endogenous or systemic mononuclear phagocytes (MPs) contribute to the removal of noxious debris, the accumulation of MPs can also result in chronic inflammation and contribute to AMD progression. In addition, direct communication and indirect molecular signaling between MPs and the RPE may promote RPE cell death, choroidal neovascularization and fibrotic scarring that occur in late AMD. In this review, we explore how the RPE and innate immune cells maintain retinal homeostasis, and detail how RPE dysfunction and aberrant immune cell recruitment contribute to AMD pathogenesis. Evidence from AMD patients will be discussed in conjunction with data from preclinical models, to shed light on future therapeutic targets for the treatment of AMD.
Collapse
|
10
|
Panayotacopoulou MT, Papageorgiou I, Pagida M, Katsogridaki AE, Chrysanthou-Piterou M, Valous NA, Halama N, Patsouris E, Konstantinidou AE. Microglia Activation in the Midbrain of the Human Neonate: The Effect of Perinatal Hypoxic-Ischemic Injury. J Neuropathol Exp Neurol 2022; 81:208-224. [PMID: 35092294 DOI: 10.1093/jnen/nlab135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Perinatal hypoxia-ischemia (PHI) is a major risk factor for the development of neuropsychiatric deficits later in life. We previously reported that after prolonged PHI, the dopaminergic neurons of the human neonate showed a dramatic reduction of tyrosine hydroxylase (TH) in the substantia nigra, without important signs of neuronal degeneration despite the significant reduction in their cell size. Since microglia activation could precede neuronal death, we now investigated 2 microglia activation markers, ionized calcium-binding adapter molecule 1 (Iba1), and the phagocytosis marker Cd68. The highest Iba1 immunoreactivity was found in neonates with neuropathological lesions of severe/abrupt PHI, while the lowest in subjects with moderate/prolonged or older PHI. Subjects with very severe/prolonged or chronic PHI showed an increased Iba1 expression and very activated microglial morphology. Heavy attachment of microglia on TH neurons and remarkable expression of Cd68 were also observed indicating phagocytosis in this group. Females appear to express more Iba1 than males, suggesting a gender difference in microglia maturation and immune reactivity after PHI insult. PHI-induced microglial "priming" during the sensitive for brain development perinatal/neonatal period, in combination with genetic or other epigenetic factors, could predispose the survivors to neuropsychiatric disorders later in life, possibly through a sexually dimorphic way.
Collapse
Affiliation(s)
- Maria T Panayotacopoulou
- From the Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P).,University Mental Health, Neurosciences and Precision Medicine Research Institute "Kostas Stefanis", National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P, EP)
| | - Ismini Papageorgiou
- Institute for Diagnostic and Interventional Radiology, University Hospital of Jena, Jena, Germany (IP).,Institute of Radiology, Südharz Hospital Nordhausen, Nordhausen, Germany (IP)
| | - Marianna Pagida
- From the Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P).,University Mental Health, Neurosciences and Precision Medicine Research Institute "Kostas Stefanis", National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P, EP)
| | - Alexandra E Katsogridaki
- From the Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P).,University Mental Health, Neurosciences and Precision Medicine Research Institute "Kostas Stefanis", National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P, EP)
| | - Margarita Chrysanthou-Piterou
- From the Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P).,University Mental Health, Neurosciences and Precision Medicine Research Institute "Kostas Stefanis", National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P, EP)
| | - Nektarios A Valous
- Applied Tumor Immunity Clinical Cooperation Unit, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany (NAV)
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Heidelberg, Germany (NH).,Division of Translational Immunotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany (NH)
| | - Efstratios Patsouris
- University Mental Health, Neurosciences and Precision Medicine Research Institute "Kostas Stefanis", National and Kapodistrian University of Athens, Athens, Greece (MTP, MP, AEK, MC-P, EP).,1st Department of Pathology, National and Kapodistrian University of Athens, Athens, Greece (EP, AEK)
| | | |
Collapse
|
11
|
Alonazi M, Ben Bacha A, Al Suhaibani A, Almnaizel AT, Aloudah HS, El-Ansary A. Psychobiotics improve propionic acid-induced neuroinflammation in juvenile rats, rodent model of autism. Transl Neurosci 2022; 13:292-300. [PMID: 36133749 PMCID: PMC9462542 DOI: 10.1515/tnsci-2022-0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 11/15/2022] Open
Abstract
This study aimed to evaluate the protective and therapeutic potency of bee pollen and probiotic mixture on brain intoxication caused by propionic acid (PPA) in juvenile rats. Five groups of six animals each, were used: the control group only receiving phosphate-buffered saline; the bee pollen and probiotic-treated group receiving a combination of an equal quantity of bee pollen and probiotic (0.2 kg/kg body weight); the PPA group being treated for 3 days with an oral neurotoxic dose of PPA (0.25 kg/kg body weight); the protective and therapeutic groups receiving bee pollen and probiotic mixture treatment right before and after the neurotoxic dose of PPA, respectively. The levels of interleukin (IL)-1ß, IL-6, IL-8, IL-10, IL-12, tumor necrosis factor α, and interferon γ (IFN-γ) were investigated to evaluate the neuroinflammatory responses in brain tissues from different animal groups. The much higher IL-1β, IL-8, and IFN-γ, as pro-inflammatory cytokines (P < 0.001), together with much lower IL-10, as anti-inflammatory cytokine (P < 0.001) compared to controls clearly demonstrated the neurotoxic effects of PPA. Interestingly, the mixture of bee pollen and probiotics was effective in alleviating PPA neurotoxic effects in both therapeutic and protective groups demonstrating highly significant changes in IL-1β, IL-8, IL-10, and IFN-γ levels together with non-significant reduction in IL-6 levels compared to PPA-treated rats. Overall, our findings demonstrated a new approach to the beneficial use of psychobiotics presenting as bee pollen and probiotic combination in neuroinflammation through cytokine changes as a possible role of glial cells in gut–brain axis.
Collapse
Affiliation(s)
- Mona Alonazi
- Biochemistry Department, Science College, King Saud University, Riyadh 11495, Saudi Arabia
| | - Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, P.O Box 22452, Riyadh 11495, Saudi Arabia.,Laboratory of Plant Biotechnology Applied to Crop Improvement, Faculty of Science of Sfax, University of Sfax, Sfax, Tunisia
| | - Anwar Al Suhaibani
- Biochemistry Department, Science College, King Saud University, Riyadh 11495, Saudi Arabia
| | - Ahmad T Almnaizel
- Experimental Surgery and Animal Laboratory, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hisham S Aloudah
- Experimental Surgery and Animal Laboratory, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Huisman Y, Uphoff K, Berger M, Dobrindt U, Schelhaas M, Zobel T, Bussmann J, van Impel A, Schulte-Merker S. Meningeal lymphatic endothelial cells fulfill scavenger endothelial cell function and cooperate with microglia in waste removal from the brain. Glia 2021; 70:35-49. [PMID: 34487573 DOI: 10.1002/glia.24081] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022]
Abstract
Brain lymphatic endothelial cells (BLECs) constitute a group of loosely connected endothelial cells that reside within the meningeal layer of the zebrafish brain without forming a vascular tubular system. BLECs have been shown to readily endocytose extracellular cargo molecules from the brain parenchyma, however, their functional relevance in relation to microglia remains enigmatic. We here compare their functional uptake efficiency for several macromolecules and bacterial components with microglia in a qualitative and quantitative manner in 5-day-old zebrafish embryos. We find BLECs to be significantly more effective in the uptake of proteins, polysaccharides and virus particles as compared to microglia, while larger particles like bacteria are only ingested by microglia but not by BLECs, implying a clear distribution of tasks between the two cell types in the brain area. In addition, we compare BLECs to the recently discovered scavenger endothelial cells (SECs) of the cardinal vein and find them to accept an identical set of substrate molecules. Our data identifies BLECs as the first brain-associated SEC population in vertebrates, and demonstrates that BLECs cooperate with microglia to remove particle waste from the brain.
Collapse
Affiliation(s)
- Yvonne Huisman
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Katharina Uphoff
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany
| | | | | | - Mario Schelhaas
- Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany.,Institute of Cellular Virology, ZMBE, Münster, Germany
| | - Thomas Zobel
- Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany.,Imaging Network, Cells in Motion Interfaculty Centre, WWU Münster, Germany
| | - Jeroen Bussmann
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands
| | - Andreas van Impel
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| |
Collapse
|
13
|
Saito ER, Miller JB, Harari O, Cruchaga C, Mihindukulasuriya KA, Kauwe JSK, Bikman BT. Alzheimer's disease alters oligodendrocytic glycolytic and ketolytic gene expression. Alzheimers Dement 2021; 17:1474-1486. [PMID: 33650792 PMCID: PMC8410881 DOI: 10.1002/alz.12310] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/05/2021] [Accepted: 01/17/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Sporadic Alzheimer's disease (AD) is strongly correlated with impaired brain glucose metabolism, which may affect AD onset and progression. Ketolysis has been suggested as an alternative pathway to fuel the brain. METHODS RNA-seq profiles of post mortem AD brains were used to determine whether dysfunctional AD brain metabolism can be determined by impairments in glycolytic and ketolytic gene expression. Data were obtained from the Knight Alzheimer's Disease Research Center (62 cases; 13 controls), Mount Sinai Brain Bank (110 cases; 44 controls), and the Mayo Clinic Brain Bank (80 cases; 76 controls), and were normalized to cell type: astrocytes, microglia, neurons, oligodendrocytes. RESULTS In oligodendrocytes, both glycolytic and ketolytic pathways were significantly impaired in AD brains. Ketolytic gene expression was not significantly altered in neurons, astrocytes, and microglia. DISCUSSION Oligodendrocytes may contribute to brain hypometabolism observed in AD. These results are suggestive of a potential link between hypometabolism and dysmyelination in disease physiology. Additionally, ketones may be therapeutic in AD due to their ability to fuel neurons despite impaired glycolytic metabolism.
Collapse
Affiliation(s)
- Erin R. Saito
- Department of Physiology and Developmental BiologyBrigham Young UniversityProvoUtahUSA
| | | | - Oscar Harari
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
- Hope Center for Neurological DisordersWashington University School of MedicineSt. LouisMissouriUSA
- NeuroGenomics and InformaticsWashington University School of MedicineSt. LouisMissouriUSA
| | - Kathie A. Mihindukulasuriya
- The Edison Family Center for Genome Sciences and Systems BiologyPathology and ImmunologyWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Benjamin T. Bikman
- Department of Physiology and Developmental BiologyBrigham Young UniversityProvoUtahUSA
| |
Collapse
|
14
|
Franco R, Lillo A, Rivas-Santisteban R, Reyes-Resina I, Navarro G. Microglial Adenosine Receptors: From Preconditioning to Modulating the M1/M2 Balance in Activated Cells. Cells 2021; 10:1124. [PMID: 34066933 PMCID: PMC8148598 DOI: 10.3390/cells10051124] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal survival depends on the glia, that is, on the astroglial and microglial support. Neurons die and microglia are activated not only in neurodegenerative diseases but also in physiological aging. Activated microglia, once considered harmful, express two main phenotypes: the pro-inflammatory or M1, and the neuroprotective or M2. When neuroinflammation, i.e., microglial activation occurs, it is important to achieve a good M1/M2 balance, i.e., at some point M1 microglia must be skewed into M2 cells to impede chronic inflammation and to afford neuronal survival. G protein-coupled receptors in general and adenosine receptors in particular are potential targets for increasing the number of M2 cells. This article describes the mechanisms underlying microglial activation and analyzes whether these cells exposed to a first damaging event may be ready to be preconditioned to better react to exposure to more damaging events. Adenosine receptors are relevant due to their participation in preconditioning. They can also be overexpressed in activated microglial cells. The potential of adenosine receptors and complexes formed by adenosine receptors and cannabinoids as therapeutic targets to provide microglia-mediated neuroprotection is here discussed.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| | - Rafael Rivas-Santisteban
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| |
Collapse
|
15
|
Wei Y, Chen T, Bosco DB, Xie M, Zheng J, Dheer A, Ying Y, Wu Q, Lennon VA, Wu LJ. The complement C3-C3aR pathway mediates microglia-astrocyte interaction following status epilepticus. Glia 2021; 69:1155-1169. [PMID: 33314324 PMCID: PMC7936954 DOI: 10.1002/glia.23955] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022]
Abstract
Gliosis is a histopathological characteristic of epilepsy that comprises activated microglia and astrocytes. It is unclear whether or how crosstalk occurs between microglia and astrocytes in the evolution of epilepsy. Here, we report in a mouse model of status epilepticus, induced by intracerebroventricular injection of kainic acid (KA), sequential activation of microglia and astrocytes and their close spatial interaction in the hippocampal CA3 region. Microglial ablation reduced astrocyte activation and their upregulation of complement C3. When compared to wild-type mice, both C3-/- and C3aR-/- mice had significantly less microglia-astrocyte interaction in response to KA-induced status epilepticus. Additionally, KA-injected C3-/- mice had significantly less histochemical evidence of neurodegeneration. The results suggest that the C3-C3aR pathway contributes to KA-induced neurodegeneration by mediating microglia-astrocyte communication. The C3-C3aR pathway may prove to be a potential therapeutic target for epilepsy treatment.
Collapse
Affiliation(s)
- Yujia Wei
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neurosurgery, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yanlu Ying
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Qian Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Vanda A. Lennon
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
16
|
Abstract
Chromenes and their derivatives have been considered as an important class of oxygen-containing heterocycles. There has been an increasing interest in the study of chromenes due to their biological activity. Herein, the structural, electronic, and vibrational properties of a chromene derivative, entitled 2‐amino‐5‐oxo‐4‐phenyl‐4,5‐dihydropyrano[3,2‐c]chromene‐3‐carbonitrile and abbreviated as Chrom-D, have been reported. The FT-IR, UV-vis, and 1H-NMR and 13C-NMR chemical shifts’ measurements were recorded. The molecular geometry and the vibrational frequencies are computed in the frame of density functional theory at the B3LYP/6-311++G(d,p) level of theory. The noncovalent interactions in the crystal lattice which are responsible to the 3D crystal structure of Chrom-D are investigated based on Hirshfeld surfaces and topological reduced density gradient (RDG) analysis. Molecular electrostatic potential surface, Mulliken charges, and Fukui functions are computed in order to find out the electrophilic and nucleophilic sites. The electronic properties of the title compound have been studied based on the TD-DFT calculations. Finally, Chrom-D has been evaluated as a multifunctional agent against Alzheimer’s disease (AD).
Collapse
|
17
|
Smith WJ, Cedeño DL, Thomas SM, Kelley CA, Vetri F, Vallejo R. Modulation of microglial activation states by spinal cord stimulation in an animal model of neuropathic pain: Comparing high rate, low rate, and differential target multiplexed programming. Mol Pain 2021; 17:1744806921999013. [PMID: 33626981 PMCID: PMC7925954 DOI: 10.1177/1744806921999013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While numerous studies and patient experiences have demonstrated the efficacy of spinal cord stimulation as a treatment for chronic neuropathic pain, the exact mechanism underlying this therapy is still uncertain. Recent studies highlighting the importance of microglial cells in chronic pain and characterizing microglial activation transcriptomes have created a focus on microglia in pain research. Our group has investigated the modulation of gene expression in neurons and glial cells after spinal cord stimulation (SCS), specifically focusing on transcriptomic changes induced by varying SCS stimulation parameters. Previous work showed that, in rodents subjected to the spared nerve injury (SNI) model of neuropathic pain, a differential target multiplexed programming (DTMP) approach provided significantly better relief of pain-like behavior compared to high rate (HRP) and low rate programming (LRP). While these studies demonstrated the importance of transcriptomic changes in SCS mechanism of action, they did not specifically address the role of SCS in microglial activation. The data presented herein utilizes microglia-specific activation transcriptomes to further understand how an SNI model of chronic pain and subsequent continuous SCS treatment with either DTMP, HRP, or LRP affects microglial activation. Genes for each activation transcriptome were identified within our dataset and gene expression levels were compared with that of healthy animals, naïve to injury and interventional procedures. Pearson correlations indicated that DTMP yields the highest significant correlations to expression levels found in the healthy animals across all microglial activation transcriptomes. In contrast, HRP or LRP yielded weak or very weak correlations for these transcriptomes. This work demonstrates that chronic pain and subsequent SCS treatments can modulate microglial activation transcriptomes, supporting previous research on microglia in chronic pain. Furthermore, this study provides evidence that DTMP is more effective than HRP and LRP at modulating microglial transcriptomes, offering potential insight into the therapeutic efficacy of DTMP.
Collapse
Affiliation(s)
- William J Smith
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - David L Cedeño
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | - Samuel M Thomas
- College of Osteopathic Medicine, Des Moines University, Des Moines, IA, USA
| | - Courtney A Kelley
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA
| | | | - Ricardo Vallejo
- Research and Development, Lumbrera LLC, Bloomington, IL, USA.,Department of Psychology, Illinois Wesleyan University, Bloomington, IL, USA.,National Spine and Pain Centers, Bloomington, IL, USA
| |
Collapse
|
18
|
Ceruti S. From astrocytes to satellite glial cells and back: A 25 year-long journey through the purinergic modulation of glial functions in pain and more. Biochem Pharmacol 2020; 187:114397. [PMID: 33382970 DOI: 10.1016/j.bcp.2020.114397] [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: 10/22/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Fundamental progresses have been made in pain research with a comprehensive understanding of the neuronal pathways which convey painful sensations from the periphery and viscera to the central nervous system and of the descending modulating pathways. Nevertheless, many patients still suffer from various painful conditions, which are often associated to other primary pathologies, and get no or poor relief from available painkillers. Thus, the interest of many researchers has concentrated on new and promising cellular targets and biochemical pathways. This is the case of glia cells, both in the peripheral and in the central nervous system, and of purinergic receptors. Starting from many intuitions and hypotheses raised by Prof. Geoffrey Burnstock, data have accumulated which clearly highlight the fundamental role exerted by several nucleotide and nucleoside receptors in the modulation of glial cell reaction to pain triggers and of their cross-talk with sensory neurons which significantly contributes to the transition from acute to chronic pain. The purinergic system has therefore become an appealing pharmacological target in pain research, also based on the quite unexpected discovery that purines are involved in ancient analgesic techniques such as acupuncture. A more in-depth understanding of the complex and intricated purine-orchestrated scenario in pain conditions will hopefully lead to the identification and clinical development of new and effective analgesics.
Collapse
Affiliation(s)
- Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milan, Italy.
| |
Collapse
|
19
|
Ferreira-Silva J, Aires ID, Boia R, Ambrósio AF, Santiago AR. Activation of Adenosine A 3 Receptor Inhibits Microglia Reactivity Elicited by Elevated Pressure. Int J Mol Sci 2020; 21:ijms21197218. [PMID: 33007835 PMCID: PMC7582754 DOI: 10.3390/ijms21197218] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 01/22/2023] Open
Abstract
Glaucoma is a progressive chronic retinal degenerative disease and a leading cause of global irreversible blindness, characterized by optic nerve damage and retinal ganglion cell (RGC) death. Elevated intraocular pressure (IOP) is a main risk factor of glaucoma. Neuroinflammation plays an important role in glaucoma. We have been demonstrating that elevated pressure triggers microglia reactivity that contribute to the loss of RGCs. Adenosine, acting on adenosine receptors, is a crucial modulator of microglia phenotype. Microglia express all adenosine receptors. Previously, we demonstrated that the activation of adenosine A3 receptor (A3R) affords protection to the retina, including RGCs, unveiling the possibility for a new strategy for glaucoma treatment. Since microglial cells express A3R, we now studied the ability of a selective A3R agonist (2-Cl-IB-MECA) in controlling microglia reactivity induced by elevated hydrostatic pressure (EHP), used to mimic elevated IOP. The activation of A3R reduced EHP-induced inducible nitric oxide synthase (iNOS) expression, microglia migration and phagocytosis in BV-2 cells. In retinal microglia, proliferation and phagocytosis elicited by EHP were also decreased by A3R activation. This work demonstrates that 2-Cl-IB-MECA, the selective agonist of A3R, is able to hinder microglia reactivity, suggesting that A3R agonists could afford protection against glaucomatous degeneration through the control of neuroinflammation.
Collapse
Affiliation(s)
- Joana Ferreira-Silva
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.F.-S.); (I.D.A.); (R.B.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Inês D. Aires
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.F.-S.); (I.D.A.); (R.B.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Raquel Boia
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.F.-S.); (I.D.A.); (R.B.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.F.-S.); (I.D.A.); (R.B.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-548 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, 3000-548 Coimbra, Portugal
| | - Ana Raquel Santiago
- Coimbra Institute for Clinical and Biomedical Research (ICBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (J.F.-S.); (I.D.A.); (R.B.); (A.F.A.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-548 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image, 3000-548 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239480226
| |
Collapse
|
20
|
Retinal ischemia triggers early microglia activation in the optic nerve followed by neurofilament degeneration. Exp Eye Res 2020; 198:108133. [PMID: 32645332 DOI: 10.1016/j.exer.2020.108133] [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: 02/07/2020] [Revised: 05/14/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Retinal ischemia leads to an early severe damage of the retina and thus plays an important role in eye diseases such as angle-closure glaucoma or retinal vascular occlusion. In retinal diseases, there is common sense about the affection of the optic nerve by ischemic injury. However, the exact dynamic processes of this optic nerve degeneration are mainly unclear. In this study, retinal ischemia was induced in one eye of Brown-Norway rats by raising the intraocular pressure 60 min to 140 mmHg followed by natural reperfusion. Optic nerves were analyzed at six different points in time: 2, 6, 12, and 24 h as well as 3 and 7 days after ischemic injury. Cell infiltration and moreover signs of tissue demyelination and dissolution were noticed in optic nerves 7 days after ischemia (hematoxylin & eosin: p < 0.001, luxol fast blue: p = 0.04). Although microglial activation was verified already from 12 h on after ischemia (p = 0.030), the beginning of a structural degeneration of the neurofilament was seen at 3 days (p = 0.02). Interestingly, proliferative microglia were present later on (7 days: p = 0.017). At this point, the number of total microglia was also increased in ischemic nerves (p = 0.003). Concluding, our data indicate that not only retinal tissue is affected by an ischemia, the optic nerve also demonstrates progressive damage. Interestingly, a microglia activation was noted days before structural damage became visible.
Collapse
|
21
|
Do HTT, Bui BP, Sim S, Jung JK, Lee H, Cho J. Anti-Inflammatory and Anti-Migratory Activities of Isoquinoline-1-Carboxamide Derivatives in LPS-Treated BV2 Microglial Cells via Inhibition of MAPKs/NF-κB Pathway. Int J Mol Sci 2020; 21:ijms21072319. [PMID: 32230861 PMCID: PMC7177615 DOI: 10.3390/ijms21072319] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022] Open
Abstract
Eleven novel isoquinoline-1-carboxamides (HSR1101~1111) were synthesized and evaluated for their effects on lipopolysaccharide (LPS)-induced production of pro-inflammatory mediators and cell migration in BV2 microglial cells. Three compounds (HSR1101~1103) exhibited the most potent suppression of LPS-induced pro-inflammatory mediators, including interleukin (IL)-6, tumor necrosis factor-alpha, and nitric oxide (NO), without significant cytotoxicity. Among them, only N-(2-hydroxyphenyl) isoquinoline-1-carboxamide (HSR1101) was found to reverse LPS-suppressed anti-inflammatory cytokine IL-10, so it was selected for further characterization. HSR1101 attenuated LPS-induced expression of inducible NO synthase and cyclooxygenase-2. Particularly, HSR1101 abated LPS-induced nuclear translocation of NF-κB through inhibition of IκB phosphorylation. Furthermore, HSR1101 inhibited LPS-induced cell migration and phosphorylation of mitogen-activated protein kinases (MAPKs) including extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38 MAPK. The specific MAPK inhibitors, U0126, SP600125, and SB203580, suppressed LPS-stimulated pro-inflammatory mediators, cell migration, and NF-κB nuclear translocation, indicating that MAPKs may be the upstream kinase of NF-κB signaling. Collectively, these results demonstrate that HSR1101 is a potent and promising compound suppressing LPS-induced inflammation and cell migration in BV2 microglial cells, and that inhibition of the MAPKs/NF-κB pathway mediates its anti-inflammatory and anti-migratory effects. Based on our findings, HSR1101 may have beneficial impacts on various neurodegenerative disorders associated with neuroinflammation and microglial activation.
Collapse
Affiliation(s)
- Ha Thi Thu Do
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (B.P.B.)
| | - Bich Phuong Bui
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (B.P.B.)
| | - Seongrak Sim
- College of Pharmacy, Chungbuk National University, Osong, Cheongju 28160, Korea; (S.S.); (J.-K.J.)
| | - Jae-Kyung Jung
- College of Pharmacy, Chungbuk National University, Osong, Cheongju 28160, Korea; (S.S.); (J.-K.J.)
| | - Heesoon Lee
- College of Pharmacy, Chungbuk National University, Osong, Cheongju 28160, Korea; (S.S.); (J.-K.J.)
- Correspondence: (H.L.); (J.C.)
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (B.P.B.)
- Correspondence: (H.L.); (J.C.)
| |
Collapse
|
22
|
Propionic acid induced behavioural effects of relevance to autism spectrum disorder evaluated in the hole board test with rats. Prog Neuropsychopharmacol Biol Psychiatry 2020; 97:109794. [PMID: 31639413 DOI: 10.1016/j.pnpbp.2019.109794] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 12/20/2022]
Abstract
Autism spectrum disorders (ASD) are a set of neurodevelopmental disorders characterized by abnormal social interactions, impaired language, and stereotypic and repetitive behaviours. Among genetically susceptible subpopulations, gut and dietary influences may play a role in etiology. Propionic acid (PPA), produced by enteric gut bacteria, crosses both the gut-blood and the blood-brain barrier. Previous research has demonstrated that repeated intracerebroventricular (ICV) infusions of PPA in adult rats produce behavioural and neuropathological changes similar to those seen in ASD patients, including hyperactivity, stereotypy, and repetitive movements. The current study examined dose and time related changes of exploratory and repetitive behaviours with the use of the hole-board task. Adult male Long-Evans rats received ICV infusions twice a day, 4 h apart, of either buffered PPA (low dose 0.052 M or high dose 0.26 M, pH 7.5, 4 μL/infusion) or phosphate buffered saline (PBS, 0.1 M) for 7 consecutive days. Locomotor activity and hole-poke behaviour were recorded daily in an automated open field apparatus (Versamax), equipped with 16 open wells, for 30 min immediately after the second infusion. In a dose dependent manner PPA infused rats displayed significantly more locomotor activity, stereotypic behaviour and nose-pokes than PBS infused rats. Low-dose PPA animals showed locomotor activity levels similar to those of PBS animals at the start of the infusion schedule, but gradually increased to levels comparable to those of high-dose PPA animals by the end of the infusion schedule, demonstrating a dose and time dependent effect of the PPA treatments.
Collapse
|
23
|
Bui BP, Oh Y, Lee H, Cho J. Inhibition of inflammatory mediators and cell migration by 1,2,3,4-tetrahydroquinoline derivatives in LPS-stimulated BV2 microglial cells via suppression of NF-κB and JNK pathway. Int Immunopharmacol 2020; 80:106231. [PMID: 32007708 DOI: 10.1016/j.intimp.2020.106231] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/31/2019] [Accepted: 01/19/2020] [Indexed: 12/16/2022]
Abstract
Novel 1,2,3,4-tetrahydroquinoline derivatives with N-alkanoyl, N-benzoyl, or chlorobenzoyl substituents were designed and synthesized to inhibit nuclear factor-kappa B (NF-κB) known to be involved in the regulation of many immune and inflammatory responses. These compounds have been previously reported to inhibit NF-κB transcriptional activity in Raw 267.4 macrophage cells and exhibit cytotoxicities to several human cancer cell lines (Jo et al., ACS Med. Chem. Lett. 7 (2016) 385-390). Accumulating evidence indicated that NF-κB is also involved in neuroinflammation implicated in many neurodegenerative diseases. Thus, the present study investigated effects of 1,2,3,4-tetrahydroquinoline derivatives on LPS-stimulated inflammatory mediators and cell migration using BV2 microglial cells as a model. We found that seven compounds tested in this study inhibited LPS-induced pro-inflammatory mediators including interleukin-6, tumor necrosis factor-α, and nitric oxide in concentration-dependent manners. Among these compounds, ELC-D-2 exhibited the most potent inhibition without showing significant cytotoxicity. We also found that ELC-D-2 attenuated levels of LPS-induced inducible nitric oxide synthase and cyclooxygenase-2. Moreover, ELC-D-2 inhibited nuclear translocation of NF-κB by suppressing inhibitor of kappa Bα phosphorylation. Furthermore, ELC-D-2 inhibited LPS-induced activation of c-Jun N-terminal kinase (JNK), which was associated with suppression of inflammatory mediators and migration of LPS-treated BV2 cells. Collectively, our findings demonstrate that ELC-D-2 inhibits LPS-induced pro-inflammatory mediators and cell migration by suppressing NF-κB translocation and JNK phosphorylation in BV2 microglial cells. These results suggest that ELC-D-2 might have a beneficial impact on various brain disorders in which neuroinflammation involving microglial activation plays a crucial role in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Bich Phuong Bui
- College of Pharmacy, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi 10326, Republic of Korea
| | - Yeonsoo Oh
- College of Pharmacy, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi 10326, Republic of Korea
| | - Heesoon Lee
- College of Pharmacy, Chungbuk National University, 194-21 Osongsaemyung-1-ro, Osong, Chungbuk 28160, Republic of Korea.
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi 10326, Republic of Korea.
| |
Collapse
|
24
|
Aghaie CI, Hausknecht KA, Wang R, Dezfuli PH, Haj-Dahmane S, Kane CJM, Sigurdson WJ, Shen RY. Prenatal Ethanol Exposure and Postnatal Environmental Intervention Alter Dopaminergic Neuron and Microglia Morphology in the Ventral Tegmental Area During Adulthood. Alcohol Clin Exp Res 2020; 44:435-444. [PMID: 31872887 DOI: 10.1111/acer.14275] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 12/13/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Prenatal ethanol exposure (PE) impairs midbrain dopaminergic (DA) neuron function, which might contribute to various cognitive and behavioral deficits, including attention deficits and increased addiction risk, often observed in individuals with fetal alcohol spectrum disorders. Currently, the underlying mechanisms for PE-induced deficits are unclear. PE could lead to neuroinflammation by activating microglia, which play an important role in synaptic function. In the present study, we investigated PE effects on microglial activation and DA neuron density and morphology in the ventral tegmental area (VTA). Since postnatal environmental enrichment can reduce neuroinflammation and ameliorate several PE-induced behavioral deficits, we examined if a postnatal environmental intervention strategy using neonatal handling and postweaning complex housing could reverse PE effects on VTA DA neurons and microglia. METHODS Pregnant rats received 0 or 6 g/kg/d ethanol by 2 intragastric intubations on gestation days 8 to 20. After birth, rats were reared in the standard laboratory or enriched condition. Male adult rats (8 to 12 weeks old) were used for immunocytochemistry. RESULTS The results showed that PE decreased VTA DA neuron body size in standardly housed rats. Moreover, there was a significant decrease in numbers of VTA microglial branches and junctions in PE rats, suggesting morphological activation of microglia and possible neuroinflammation. The PE effects on microglia were normalized by postnatal environmental intervention, which also decreased the numbers of microglial branches and junctions in control animals, possibly via reduced stress. CONCLUSIONS Our findings show an association between PE-induced morphological activation of microglia and impaired DA neuron morphology in the VTA. Importantly, postnatal environmental intervention rescues possible PE-induced microglial activation. These data support that environmental intervention can be effective in ameliorating cognitive and behavioral deficits associated with VTA DA neuron dysfunctions, such as attention deficits and increased addiction risk.
Collapse
Affiliation(s)
- Claudia I Aghaie
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, New York
| | - Kathryn A Hausknecht
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York
| | - Ruixiang Wang
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York
| | | | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York
| | - Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, College of Medicine, the University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Wade J Sigurdson
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, New York
| | - Roh-Yu Shen
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, New York.,Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York
| |
Collapse
|
25
|
Wang T, Yan B, Lou L, Lin X, Yu T, Wu S, Lu Q, Liu W, Huang Z, Zhang M, Zhang W, Wen Z. Nlrc3-like is required for microglia maintenance in zebrafish. J Genet Genomics 2019; 46:291-299. [PMID: 31278008 DOI: 10.1016/j.jgg.2019.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 01/06/2023]
Abstract
Microglia are tissue-resident macrophages residing in the central nervous system (CNS) and play critical roles in removing cellular debris and infectious agents as well as regulating neurogenesis and neuronal activities. Yet, the molecular basis underlying the establishment of microglia pool and the maintenance of their homeostasis in the CNS remain largely undefined. Here we report the identification and characterization of a mutant zebrafish, which harbors a point mutation in the nucleotide-binding oligomerization domain (NOD) like receptor gene nlrc3-like, resulting in the loss of microglia in a temperature sensitive manner. Temperature shift assay reveals that the late onset of nlrc3-like deficiency leads to excessive microglia cell death. Further analysis shows that the excessive microglia death in nlrc3-like deficient mutants is attributed, at least in part, to aberrant activation of canonical inflammasome pathway. Our study indicates that proper regulation of inflammasome cascade is critical for the maintenance of microglia homeostasis.
Collapse
Affiliation(s)
- Tienan Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China; Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Bo Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China; Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| | - Liang Lou
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Xi Lin
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Tao Yu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Shuting Wu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Qing Lu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Liu
- Department of Developmental Biology, School of Basic Medical Sciences, South China University of Technology, Guangzhou, 510630, China
| | - Zhibin Huang
- Department of Developmental Biology, School of Basic Medical Sciences, South China University of Technology, Guangzhou, 510630, China
| | - Mingjie Zhang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China
| | - Wenqing Zhang
- Department of Developmental Biology, School of Basic Medical Sciences, South China University of Technology, Guangzhou, 510630, China.
| | - Zilong Wen
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
26
|
Green LA, Nebiolo JC, Smith CJ. Microglia exit the CNS in spinal root avulsion. PLoS Biol 2019; 17:e3000159. [PMID: 30794533 PMCID: PMC6402705 DOI: 10.1371/journal.pbio.3000159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 03/06/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Microglia are central nervous system (CNS)-resident cells. Their ability to migrate outside of the CNS, however, is not understood. Using time-lapse imaging in an obstetrical brachial plexus injury (OBPI) model, we show that microglia squeeze through the spinal boundary and emigrate to peripheral spinal roots. Although both macrophages and microglia respond, microglia are the debris-clearing cell. Once outside the CNS, microglia re-enter the spinal cord in an altered state. These peripheral nervous system (PNS)-experienced microglia can travel to distal CNS areas from the injury site, including the brain, with debris. This emigration is balanced by two mechanisms—induced emigration via N-methyl-D-aspartate receptor (NMDA) dependence and restriction via contact-dependent cellular repulsion with macrophages. These discoveries open the possibility that microglia can migrate outside of their textbook-defined regions in disease states. Microglia are normally assumed to be confined to the central nervous system (CNS), but this study shows show that after spinal root injury, microglia can exit the CNS to clear debris. Upon re-entry, the emigrated microglia are altered and can travel to distal areas such as the brain. Cells are precisely organized in specific anatomical domains to ensure normal functioning of the nervous system. One such cell type, microglia, is usually considered to be confined to the central nervous system (CNS). Using time-lapse imaging to capture microglia as they migrate, we show that their characteristic CNS-residency can be altered after spinal root injury. After such injury, the microglia exit the spinal root to the periphery, where they clear debris at the injury site and then carry that debris back into the CNS. In addition, microglia that leave the CNS after spinal root injury become distinct from those that remain within the CNS. This emigration event of microglia after injury is driven by two mechanisms—dependence on glutamatergic signaling that induces their emigration to the injury and interactions with macrophages that prevent their ectopic exit from the spinal cord. Together, these discoveries raise the possibility that microglia could override their CNS-residency in certain disease contexts.
Collapse
Affiliation(s)
- Lauren A Green
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Julia C Nebiolo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
27
|
Xu D, Jin T, Zhu H, Chen H, Ofengeim D, Zou C, Mifflin L, Pan L, Amin P, Li W, Shan B, Naito MG, Meng H, Li Y, Pan H, Aron L, Adiconis X, Levin JZ, Yankner BA, Yuan J. TBK1 Suppresses RIPK1-Driven Apoptosis and Inflammation during Development and in Aging. Cell 2018; 174:1477-1491.e19. [PMID: 30146158 PMCID: PMC6128749 DOI: 10.1016/j.cell.2018.07.041] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/28/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
Abstract
Aging is a major risk factor for both genetic and sporadic neurodegenerative disorders. However, it is unclear how aging interacts with genetic predispositions to promote neurodegeneration. Here, we investigate how partial loss of function of TBK1, a major genetic cause for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comorbidity, leads to age-dependent neurodegeneration. We show that TBK1 is an endogenous inhibitor of RIPK1 and the embryonic lethality of Tbk1-/- mice is dependent on RIPK1 kinase activity. In aging human brains, another endogenous RIPK1 inhibitor, TAK1, exhibits a marked decrease in expression. We show that in Tbk1+/- mice, the reduced myeloid TAK1 expression promotes all the key hallmarks of ALS/FTD, including neuroinflammation, TDP-43 aggregation, axonal degeneration, neuronal loss, and behavior deficits, which are blocked upon inhibition of RIPK1. Thus, aging facilitates RIPK1 activation by reducing TAK1 expression, which cooperates with genetic risk factors to promote the onset of ALS/FTD.
Collapse
Affiliation(s)
- Daichao Xu
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Taijie Jin
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Rd., Pudong, 201210 Shanghai, China; University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Hong Zhu
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Hongbo Chen
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Dimitry Ofengeim
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Chengyu Zou
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Lifeng Pan
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Rd., 200032 Shanghai, China
| | - Palak Amin
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Wanjin Li
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Rd., Pudong, 201210 Shanghai, China
| | - Masanori Gomi Naito
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | - Huyan Meng
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Rd., Pudong, 201210 Shanghai, China; University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Rd., Pudong, 201210 Shanghai, China
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Rd., Pudong, 201210 Shanghai, China
| | - Liviu Aron
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Bruce A Yankner
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA; Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Rd., Pudong, 201210 Shanghai, China.
| |
Collapse
|
28
|
Hammond TR, Robinton D, Stevens B. Microglia and the Brain: Complementary Partners in Development and Disease. Annu Rev Cell Dev Biol 2018; 34:523-544. [PMID: 30089221 DOI: 10.1146/annurev-cellbio-100616-060509] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An explosion of findings driven by powerful new technologies has expanded our understanding of microglia, the resident immune cells of the central nervous system (CNS). This wave of discoveries has fueled a growing interest in the roles that these cells play in the development of the CNS and in the neuropathology of a diverse array of disorders. In this review, we discuss the crucial roles that microglia play in shaping the brain-from their influence on neurons and glia within the developing CNS to their roles in synaptic maturation and brain wiring-as well as some of the obstacles to overcome when assessing their contributions to normal brain development. Furthermore, we examine how normal developmental functions of microglia are perturbed or remerge in neurodevelopmental and neurodegenerative disease.
Collapse
Affiliation(s)
- Timothy R Hammond
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Daisy Robinton
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Beth Stevens
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA; .,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
29
|
Lee JC, Shin BN, Cho JH, Lee TK, Kim IH, Noh Y, Kim SS, Lee HA, Kim YM, Kim H, Cho JH, Park JH, Ahn JH, Kang IJ, Hwang IK, Won MH, Shin MC. Brain ischemic preconditioning protects against moderate, not severe, transient global cerebral ischemic injury. Metab Brain Dis 2018; 33:1193-1201. [PMID: 29644488 DOI: 10.1007/s11011-018-0231-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
Ischemic preconditioning (IPC) in the brain increases ischemic tolerance to subsequent ischemic insults. In this study, we examined whether IPC protects neurons and attenuates microgliosis or not in the hippocampus following severe transient global cerebral ischemia (TCI) in gerbils. Gerbils were assigned to 8 groups; 5- and 15-min sham operated groups, 5-min and 15-min TCI operated groups, IPC plus 5- and 15-min sham operated groups, and IPC plus 5- and 15-min TCI operated groups. IPC was induced by subjecting animals to 2-min transient ischemia 1 day before 5-min TCI for a typical transient ischemia and 15-min TCI for severe transient ischemia. Neuronal damage was examined by cresyl violet staining and Fluoro-Jade B histofluorescence staining. In addition, microglial activation was examined using immunohistochemistry for Iba-1 (a marker for microglia). Delayed neuronal death and microgliosis was found in the CA1 alone in the 5-min TCI operated group at 5 days post-ischemia, and, in the 15-min TCI operated group, neuronal death and microgliosis was shown in all CA areas (CA1-3) and the dentate gyrus. IPC displayed neuroprotection and attenuated microglial activation in the 5-min TCI operated group. However, in the 15-min TCI operated group, IPC did not show neuroprotection and not attenuate microglial activation. Our present findings indicate that IPC hardly protect against severe transient cerebral ischemic injury.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Bich-Na Shin
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - In Hye Kim
- Famenity Company, Gwacheon, 13837, Republic of Korea
| | - YooHun Noh
- Famenity Company, Gwacheon, 13837, Republic of Korea
| | - Sung-Su Kim
- Famenity Company, Gwacheon, 13837, Republic of Korea
| | - Hyang-Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyeyoung Kim
- Department of Anesthesiology and Pain Medicine, Chungju Hospital, Konkuk University School of Medicine, Chungju, 27376, Republic of Korea
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, 24252, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
30
|
Moniruzzaman M, Chin YW, Cho J. HO-1 dependent antioxidant effects of ethyl acetate fraction from Physalis alkekengi fruit ameliorates scopolamine-induced cognitive impairments. Cell Stress Chaperones 2018; 23:763-772. [PMID: 29546728 PMCID: PMC6045529 DOI: 10.1007/s12192-018-0887-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/04/2018] [Accepted: 02/12/2018] [Indexed: 12/21/2022] Open
Abstract
Physalis alkekengi var. francheti is an indigenous herb well known for its anti-inflammatory, sedative, antipyretic, and expectorant properties. However, the information regarding the impacts of P. alkekengi fruits (PAF) in modulation of oxidative stress and learning memory are still unknown. This study therefore evaluated the antioxidant properties of ethyl acetate (EA) fraction of PAF and its impacts on learning and memory. The antioxidant activities of PAF were evaluated in LPS-induced BV2 microglial cells. The potent EA fraction then investigated and confirmed for its involvement of HO-1 pathway using hemin (HO-1 inducer) and ZnPP (HO-1 inhibitor) through Western blotting, DCFH-DA, and/or Griess assay. The involvements of PI3K/Akt, MEK, and p38 MAPK also investigated. Furthermore, we applied EA fraction to the animals at 100 and 200 mg/kg doses to check if the extract could improve scopolamine-induced memory deficits in passive avoidance and elevated plus maze tests. Our results demonstrated that the fractions from PAF significantly inhibited the generation of intracellular reactive oxygen species (ROS) induced by LPS in concentration-dependent manners. In comparison to other fractions, the EA fraction exhibited potent effect in suppressing intracellular ROS generation. Besides, EA fraction also induced the expression of HO-1 in time- and concentration-dependent manners. ZnPP significantly reversed the suppressive effect of EA fraction on LPS-induced ROS generation and NO production, which confirm the involvement of HO-1 signaling in EA-fraction-mediated antioxidant activities. Consistently, blocking of PI3K/Akt, MEK, and p38 MAPK pathways by PAF-EA suppressed the production of intracellular ROS, indicating their potential participation. In addition, one of the major constituents of EA fraction, luteolin-7-O-β-D-glucoside, also demonstrated HO-1-dependent antioxidant effects in BV2 cells. Further, the EA fraction significantly (p < 0.05) improves scopolamine-induced memory deficits in mice. Taken together, our findings highlight the antioxidant effects of EA fraction of PAF which may be beneficial in treatment of different neurodegenerative diseases associated with free radicals.
Collapse
Affiliation(s)
- Md Moniruzzaman
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea.
- Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, QLD, 4102, Australia.
| | - Young-Won Chin
- College of Pharmacy and BK-Plus Team, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
31
|
Mehrabi M, Amini F, Mehrabi S. Active Role of the Necrotic Zone in Desensitization of Hypoxic Macrophages and Regulation of CSC-Fate: A hypothesis. Front Oncol 2018; 8:235. [PMID: 29988496 PMCID: PMC6026632 DOI: 10.3389/fonc.2018.00235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/11/2018] [Indexed: 01/30/2023] Open
Abstract
Fast-proliferating cancer cells in the hypoxic region face a shortage of oxygen and nutrients, undergo necrotic cell death, and release numerous signaling components. Hypoxia-induced chemo-attractants signal for macrophages/monocytes to clear debris and return the system to steady state. Accordingly, macrophages arrange into pre-necrotic positions, where they are continuously exposed to stress signals. It can thus be hypothesized that gradual alteration of gene expression in macrophages eventually turns off their phagocytic machinery. Uncleared cell corpses within the hypoxic region potentially provide a rich source of building blocks for anaerobic metabolism of cancer stem cells via macropinocytosis, and are conceivably implicated in tumor progression and invasion.
Collapse
Affiliation(s)
| | - Fatemeh Amini
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, United Kingdom
| | - Shima Mehrabi
- Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Prenylated Flavonoids from Cudrania tricuspidata Suppress Lipopolysaccharide-Induced Neuroinflammatory Activities in BV2 Microglial Cells. Int J Mol Sci 2016; 17:255. [PMID: 26907256 PMCID: PMC4783984 DOI: 10.3390/ijms17020255] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 01/12/2016] [Accepted: 02/05/2016] [Indexed: 11/16/2022] Open
Abstract
In Korea and China, Cudrania tricuspidata Bureau (Moraceae) is an important traditional medicinal plant used to treat lumbago, hemoptysis, and contusions. The C. tricuspidata methanol extract suppressed both production of NO and PGE2 in BV2 microglial cells. Cudraflavanone D (1), isolated from this extract, remarkably suppressed the protein expression of inducible NO synthase and cyclooxygenase-2, and decreased the levels of NO and PGE2 in BV2 microglial cells exposed to lipopolysaccharide. Cudraflavanone D (1) also decreased IL-6, TNF-α, IL-12, and IL-1β production, blocked nuclear translocation of NF-κB heterodimers (p50 and p65) by interrupting the degradation and phosphorylation of inhibitor of IκB-α, and inhibited NF-κB binding. In addition, cudraflavanone D (1) suppressed the phosphorylation of c-Jun N-terminal kinase (JNK) and p38 MAPK pathways. This study indicated that cudraflavanone D (1) can be a potential drug candidate for the cure of neuroinflammation.
Collapse
|
33
|
Zhang S, Shao SY, Song XY, Xia CY, Yang YN, Zhang PC, Chen NH. Protective effects of Forsythia suspense extract with antioxidant and anti-inflammatory properties in a model of rotenone induced neurotoxicity. Neurotoxicology 2016; 52:72-83. [DOI: 10.1016/j.neuro.2015.09.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 12/21/2022]
|
34
|
de Gracia P, Gallego BI, Rojas B, Ramírez AI, de Hoz R, Salazar JJ, Triviño A, Ramírez JM. Automatic Counting of Microglial Cells in Healthy and Glaucomatous Mouse Retinas. PLoS One 2015; 10:e0143278. [PMID: 26580208 PMCID: PMC4651327 DOI: 10.1371/journal.pone.0143278] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/02/2015] [Indexed: 12/30/2022] Open
Abstract
Proliferation of microglial cells has been considered a sign of glial activation and a hallmark of ongoing neurodegenerative diseases. Microglia activation is analyzed in animal models of different eye diseases. Numerous retinal samples are required for each of these studies to obtain relevant data of statistical significance. Because manual quantification of microglial cells is time consuming, the aim of this study was develop an algorithm for automatic identification of retinal microglia. Two groups of adult male Swiss mice were used: age-matched controls (naïve, n = 6) and mice subjected to unilateral laser-induced ocular hypertension (lasered; n = 9). In the latter group, both hypertensive eyes and contralateral untreated retinas were analyzed. Retinal whole mounts were immunostained with anti Iba-1 for detecting microglial cell populations. A new algorithm was developed in MATLAB for microglial quantification; it enabled the quantification of microglial cells in the inner and outer plexiform layers and evaluates the area of the retina occupied by Iba-1+ microglia in the nerve fiber-ganglion cell layer. The automatic method was applied to a set of 6,000 images. To validate the algorithm, mouse retinas were evaluated both manually and computationally; the program correctly assessed the number of cells (Pearson correlation R = 0.94 and R = 0.98 for the inner and outer plexiform layers respectively). Statistically significant differences in glial cell number were found between naïve, lasered eyes and contralateral eyes (P<0.05, naïve versus contralateral eyes; P<0.001, naïve versus lasered eyes and contralateral versus lasered eyes). The algorithm developed is a reliable and fast tool that can evaluate the number of microglial cells in naïve mouse retinas and in retinas exhibiting proliferation. The implementation of this new automatic method can enable faster quantification of microglial cells in retinal pathologies.
Collapse
Affiliation(s)
- Pablo de Gracia
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, United States of America
- * E-mail:
| | - Beatriz I. Gallego
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - Blanca Rojas
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Óptica y Optometría, Universidad Complutense de Madrid, Madrid, Spain
| | - Alberto Triviño
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Xavier AL, Menezes JRL, Goldman SA, Nedergaard M. Fine-tuning the central nervous system: microglial modelling of cells and synapses. Philos Trans R Soc Lond B Biol Sci 2015; 369:20130593. [PMID: 25225087 DOI: 10.1098/rstb.2013.0593] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Microglia constitute as much as 10-15% of all cells in the mammalian central nervous system (CNS) and are the only glial cells that do not arise from the neuroectoderm. As the principal CNS immune cells, microglial cells represent the first line of defence in response to exogenous threats. Past studies have largely been dedicated to defining the complex immune functions of microglial cells. However, our understanding of the roles of microglia has expanded radically over the past years. It is now clear that microglia are critically involved in shaping neural circuits in both the developing and adult CNS, and in modulating synaptic transmission in the adult brain. Intriguingly, microglial cells appear to use the same sets of tools, including cytokine and chemokine release as well as phagocytosis, whether modulating neural function or mediating the brain's innate immune responses. This review will discuss recent developments that have broadened our views of neuro-glial signalling to include the contribution of microglial cells.
Collapse
Affiliation(s)
- Anna L Xavier
- Laboratório de Neuroanatomia Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY 14642, USA
| | - João R L Menezes
- Laboratório de Neuroanatomia Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Steven A Goldman
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY 14642, USA
| |
Collapse
|
36
|
Morsch M, Radford R, Lee A, Don EK, Badrock AP, Hall TE, Cole NJ, Chung R. In vivo characterization of microglial engulfment of dying neurons in the zebrafish spinal cord. Front Cell Neurosci 2015; 9:321. [PMID: 26379496 PMCID: PMC4553390 DOI: 10.3389/fncel.2015.00321] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/03/2015] [Indexed: 11/13/2022] Open
Abstract
Microglia are specialized phagocytes in the vertebrate central nervous system (CNS). As the resident immune cells of the CNS they play an important role in the removal of dying neurons during both development and in several neuronal pathologies. Microglia have been shown to prevent the diffusion of damaging degradation products of dying neurons by engulfment and ingestion. Here we describe a live imaging approach that uses UV laser ablation to selectively stress and kill spinal neurons and visualize the clearance of neuronal remnants by microglia in the zebrafish spinal cord. In vivo imaging confirmed the motile nature of microglia within the uninjured spinal cord. However, selective neuronal ablation triggered rapid activation of microglia, leading to phagocytic uptake of neuronal debris by microglia within 20-30 min. This process of microglial engulfment is highly dynamic, involving the extension of processes toward the lesion site and consequently the ingestion of the dying neuron. 3D rendering analysis of time-lapse recordings revealed the formation of phagosome-like structures in the activated microglia located at the site of neuronal ablation. This real-time representation of microglial phagocytosis in the living zebrafish spinal cord provides novel opportunities to study the mechanisms of microglia-mediated neuronal clearance.
Collapse
Affiliation(s)
- Marco Morsch
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Rowan Radford
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Albert Lee
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Emily K Don
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Andrew P Badrock
- Faculty of Life Sciences, The University of Manchester Manchester, UK
| | - Thomas E Hall
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| | - Nicholas J Cole
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Roger Chung
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| |
Collapse
|
37
|
Nowlin BT, Burdo TH, Midkiff CC, Salemi M, Alvarez X, Williams KC. SIV encephalitis lesions are composed of CD163(+) macrophages present in the central nervous system during early SIV infection and SIV-positive macrophages recruited terminally with AIDS. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1649-65. [PMID: 25963554 DOI: 10.1016/j.ajpath.2015.01.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/16/2015] [Accepted: 01/30/2015] [Indexed: 10/23/2022]
Abstract
Macrophage recruitment to the central nervous system (CNS) during AIDS pathogenesis is poorly understood. We measured the accumulation of brain perivascular (CD163(+)) and inflammatory (MAC387(+)) macrophages in SIV-infected monkeys. Monocyte progenitors were 5-bromo-2'-deoxyuridine (BrdU) labeled in bone marrow, and CNS macrophages were labeled serially with fluorescent dextrans injected into the cisterna magna. MAC387(+) macrophages accumulated in the meninges and choroid plexus in early inflammation and in the perivascular space and SIV encephalitis (SIVE) lesions late. CD163(+) macrophages accumulated in the perivascular space and SIVE lesions with late inflammation. Most of the BrdU(+) cells were MAC387(+); however, CD163(+)BrdU(+) macrophages were present in the meninges and choroid plexus with AIDS. Most (81.6% ± 1.8%) of macrophages in SIVE lesions were present in the CNS before SIVE lesion formation. There was a 2.9-fold increase in SIVp28(+) macrophages entering the CNS late compared with those entering early (P < 0.05). The rate of CD163(+) macrophage recruitment to the CNS inversely correlated with time to death (P < 0.03) and increased with SIVE. In SIVE animals, soluble CD163 correlated with CD163(+) macrophage recruitment (P = 0.02). Most perivascular macrophages that comprise SIVE lesions and multinucleated giant cells are present in the CNS early, before SIVE lesions are formed. Most SIV-infected macrophages traffic to the CNS terminally with AIDS.
Collapse
Affiliation(s)
- Brian T Nowlin
- Biology Department, Boston College, Chestnut Hill, Massachusetts
| | - Tricia H Burdo
- Biology Department, Boston College, Chestnut Hill, Massachusetts
| | - Cecily C Midkiff
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University Health Science Center, Covington, Louisiana
| | - Marco Salemi
- Department of Pathology, Immunology, and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, Florida
| | - Xavier Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Tulane University Health Science Center, Covington, Louisiana
| | | |
Collapse
|
38
|
Peng J, Wang P, Ge H, Qu X, Jin X. Effects of cordycepin on the microglia-overactivation-induced impairments of growth and development of hippocampal cultured neurons. PLoS One 2015; 10:e0125902. [PMID: 25932642 PMCID: PMC4416906 DOI: 10.1371/journal.pone.0125902] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/20/2015] [Indexed: 12/24/2022] Open
Abstract
Microglial cells are normally activated in response to brain injury or immunological stimuli to protect central nervous system (CNS). However, over-activation of microglia conversely amplifies the inflammatory effects and mediates cellular degeneration, leading to the death of neurons. Recently, cordycepin, an active component found in Cordyceps militarisa known as a rare Chinese caterpillar fungus, has been reported as an effective drug for treating inflammatory diseases and cancer via unclear mechanisms. In this study, we attempted to identify the anti-inflammatory role of cordycepin and its protective effects on the impairments of neural growth and development induced by microglial over-activation. The results indicate that cordycepin could attenuate the lipopolysaccharide (LPS)-induced microglial activation, evidenced by the dramatically reduced release of TNF-α and IL-1β, as well as the down-regulation of mRNA levels of iNOS and COX-2 after cordycepin treatment. Besides, cordycepin reversed the LPS-induced activation of NF-κB pathway, resulting in anti-inflammatory effects. Furthermore, by employing the conditioned medium (CM), we found cordycepin was able to recover the impairments of neural growth and development in the primary hippocampal neurons cultured in LPS-CM, including cell viability, growth cone extension, neurite sprouting and outgrowth as well as spinogenesis. This study expands our knowledge of the anti-inflammatory function of cordycepin and paves the way for the biomedical applications of cordycepin in the therapies of neural injuries.
Collapse
Affiliation(s)
- Jie Peng
- Wuzhong Hospital, Suzhou, Jiangsu, China
| | - Ping Wang
- Wuzhong Hospital, Suzhou, Jiangsu, China
| | - Hongshan Ge
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xianqin Qu
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, NSW, Australia
- * E-mail: (XJ); (XQ)
| | - Xingliang Jin
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Sydney Centre for Regenerative and Developmental Medicine, Kolling Institute for Medical Research, Sydney Medical School, University of Sydney, NSW, Australia
- * E-mail: (XJ); (XQ)
| |
Collapse
|
39
|
Zhang H, Li F, Yang Y, Chen J, Hu X. SIRP/CD47 signaling in neurological disorders. Brain Res 2015; 1623:74-80. [PMID: 25795378 DOI: 10.1016/j.brainres.2015.03.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 11/26/2022]
Abstract
Microglia play important roles in the process of neuronal injury and recovery. Numeous surface receptors have been described to regulate microglial activation. These receptors tightly mediate normal microglial functions including cell mobility, phagocytosis, and production of inflammatory mediators or trophic factors. In recent years, significant progresses have been achieved for understanding the signaling mechanisms underlying these receptors. Their specific roles in neurological diseases have been documented. This review will focus on the signal regulatory protein (SIRP) and its ligand CD47, two surface receptors expressed on microglia and other cells in the central nervous system (CNS) such as neurons. We will discuss the involvement of SIRP/CD47 signaling in microglial activation and in the interplay between microglia and other CNS cells. Current studies reveal the importance of CD47 and SIRPα in the process of neuroinflammation in the CNS disorders. The dual and contradictory role of CD47 suggests that targeting the SIRPα/CD47 signaling may achieve different effects depending on disease stage. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Haiyue Zhang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Xianaya School of Medicine, Central South University, Changsha, Hunan, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Luhe Teaching Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Yang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Xianaya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; China-America Institute of Neuroscience, Luhe Teaching Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
40
|
Latta CH, Brothers HM, Wilcock DM. Neuroinflammation in Alzheimer's disease; A source of heterogeneity and target for personalized therapy. Neuroscience 2014; 302:103-11. [PMID: 25286385 DOI: 10.1016/j.neuroscience.2014.09.061] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/30/2022]
Abstract
Neuroinflammation has long been known as an accompanying pathology of Alzheimer's disease. Microglia surrounding amyloid plaques in the brain of Auguste D were described in the original publication of Alois Alzheimer. It is only quite recently, however, that we have a more complete appreciation for the diverse roles of neuroinflammation in neurodegenerative disorders such as Alzheimer's. While gaps in our knowledge remain, and conflicting data are abound in the field, our understanding of the complexities and heterogeneous functions of the inflammatory response in Alzheimer's is vastly improved. This review article will discuss some of the roles of neuroinflammation in Alzheimer's disease, in particular, how understanding heterogeneity in the individual inflammatory response can be used in therapeutic development and as a mechanism of personalizing our treatment of the disease.
Collapse
Affiliation(s)
- C H Latta
- University of Kentucky, Sanders-Brown Center on Aging, Department of Physiology, Lexington, KY 40536, USA; The University of Manchester, Department of Biology, Manchester M13 9PL, United Kingdom
| | - H M Brothers
- University of Kentucky, Sanders-Brown Center on Aging, Department of Physiology, Lexington, KY 40536, USA
| | - D M Wilcock
- University of Kentucky, Sanders-Brown Center on Aging, Department of Physiology, Lexington, KY 40536, USA.
| |
Collapse
|
41
|
Liu F, Zhou R, Yan H, Yin H, Wu X, Tan Y, Li L. Metabotropic glutamate receptor 5 modulates calcium oscillation and innate immune response induced by lipopolysaccharide in microglial cell. Neuroscience 2014; 281:24-34. [PMID: 25250840 DOI: 10.1016/j.neuroscience.2014.09.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/13/2014] [Accepted: 09/15/2014] [Indexed: 01/01/2023]
Abstract
Microglia, the primary immune cells in the brain, have been implicated as the predominant cells governing inflammation-mediated neuronal damage. In response to immunological challenges such as lipopolysaccharide (LPS), microglia are activated and subsequently inflammatory process is initiated as evidenced by the release of pro-inflammatory chemokines and cytokines. Here we show that Group I metabotropic glutamate receptor 5 (mGluR5) is involved in LPS-induced microglia activation. LPS triggered a similar pattern of [Ca2+]i oscillation in N9, Toll-like receptor 4 (TLR4)-mutant EOC 20, TLR4-wild-type and TLR4-deficient primary mouse microglia, suggesting that LPS-induced [Ca2+]i oscillation is independent of TLR4. The characteristics of [Ca2+]i oscillation induced by LPS are consistent with those observed in mGluR5 activation. In addition, mGluR5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) abolished LPS-induced [Ca2+]i oscillation. Immunocytochemistry demonstrated that LPS colocalizes with mGluR5 in microglia and the direct binding of LPS and mGluR5 was further validated by antibody-based fluorescence resonance energy transfer (FRET) technology. Activation of mGluR5 using a selective agonist (RS)-2-chloro-5-hydroxyphenylglycine (CHPG) significantly expanded LPS-induced nuclear factor-kappa B (NF-κB) activity and CHPG alone increased NF-κB activity as well. But, mGluR5 antagonist MTEP attenuated the actions of LPS, CHPG and the additive effect of LPS and CHPG in microglia. LPS induced tumor necrosis factor-α (TNF-α) secretion in N9 microglia, but not in TLR4-mutant EOC 20 and TLR4-deficient primary mouse microglia. CHPG reduced LPS-caused TNF-α production, but MTEP increased LPS-induced TNF-α production and blocked the effect of CHPG in N9 microglia. These data demonstrate that mGluR5 and TLR4 are two critical receptors that mediate microglia activation in response to LPS, suggesting that mGluR5 may represent a novel target for modulating microglia-dependent neuroinflammation.
Collapse
Affiliation(s)
- F Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China; Department of Infection & Immunity, Research Institute of Surgery/Daping Hospital, Third Military Medical University, Chongqing, China
| | - R Zhou
- Department of Operating Room, Children's Hospital, Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital, Chongqing Medical University, Chongqing, China
| | - H Yan
- Department of Anesthesiology, Research Institute of Surgery/Daping Hospital, Third Military Medical University, Chongqing, China
| | - H Yin
- Department of Emergency Nursing, School of Nursing, Third Military Medical University, Chongqing, China
| | - X Wu
- Department of Infection & Immunity, Research Institute of Surgery/Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y Tan
- Department of Infection & Immunity, Research Institute of Surgery/Daping Hospital, Third Military Medical University, Chongqing, China
| | - L Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China; Department of Infection & Immunity, Research Institute of Surgery/Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
42
|
Siebert JR, Conta Steencken A, Osterhout DJ. Chondroitin sulfate proteoglycans in the nervous system: inhibitors to repair. BIOMED RESEARCH INTERNATIONAL 2014; 2014:845323. [PMID: 25309928 PMCID: PMC4182688 DOI: 10.1155/2014/845323] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 12/14/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are widely expressed in the normal central nervous system, serving as guidance cues during development and modulating synaptic connections in the adult. With injury or disease, an increase in CSPG expression is commonly observed close to lesioned areas. However, these CSPG deposits form a substantial barrier to regeneration and are largely responsible for the inability to repair damage in the brain and spinal cord. This review discusses the role of CSPGs as inhibitors, the role of inflammation in stimulating CSPG expression near site of injury, and therapeutic strategies for overcoming the inhibitory effects of CSPGs and creating an environment conducive to nerve regeneration.
Collapse
Affiliation(s)
- Justin R. Siebert
- Lake Erie College of Osteopathic Medicine at Seton Hill, 20 Seton Hill Drive, Greensburg, PA 15601, USA
| | - Amanda Conta Steencken
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Donna J. Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
43
|
Rojas B, Gallego BI, Ramírez AI, Salazar JJ, de Hoz R, Valiente-Soriano FJ, Avilés-Trigueros M, Villegas-Perez MP, Vidal-Sanz M, Triviño A, Ramírez JM. Microglia in mouse retina contralateral to experimental glaucoma exhibit multiple signs of activation in all retinal layers. J Neuroinflammation 2014; 11:133. [PMID: 25064005 PMCID: PMC4128533 DOI: 10.1186/1742-2094-11-133] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/15/2014] [Indexed: 01/02/2023] Open
Abstract
Background Glaucomatous optic neuropathy, a leading cause of blindness, can progress despite control of intraocular pressure - currently the main risk factor and target for treatment. Glaucoma progression shares mechanisms with neurodegenerative disease, including microglia activation. In the present model of ocular hypertension (OHT), we have recently described morphological signs of retinal microglia activation and MHC-II upregulation in both the untreated contralateral eyes and OHT eyes. By using immunostaining, we sought to analyze and quantify additional signs of microglia activation and differences depending on the retinal layer. Methods Two groups of adult Swiss mice were used: age-matched control (naïve, n = 12), and lasered (n = 12). In the lasered animals, both OHT eyes and contralateral eyes were analyzed. Retinal whole-mounts were immunostained with antibodies against Iba-1, MHC-II, CD68, CD86, and Ym1. The Iba-1+ cell number in the plexiform layers (PL) and the photoreceptor outer segment (OS), Iba-1+ arbor area in the PL, and area of the retina occupied by Iba-1+ cells in the nerve fiber layer-ganglion cell layer (NFL-GCL) were quantified. Results The main findings in contralateral eyes and OHT eyes were: i) ameboid microglia in the NFL-GCL and OS; ii) the retraction of processes in all retinal layers; iii) a higher level of branching in PL and in the OS; iv) soma displacement to the nearest cell layers in the PL and OS; v) the reorientation of processes in the OS; vi) MHC-II upregulation in all retinal layers; vii) increased CD68 immunostaining; and viii) CD86 immunolabeling in ameboid cells. In comparison with the control group, a significant increase in the microglial number in the PL, OS, and in the area occupied by Iba-1+ cells in the NFL-GCL, and significant reduction of the arbor area in the PL. In addition, rounded Iba-1+ CD86+ cells in the NFL-GCL, OS and Ym1+ cells, and rod-like microglia in the NFL-GCL were restricted to OHT eyes. Conclusions Several quantitative and qualitative signs of microglia activation are detected both in the contralateral and OHT eyes. Such activation extended beyond the GCL, involving all retinal layers. Differences between the two eyes could help to elucidate glaucoma pathophysiology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - José M Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Facultad de Medicina, Pab VI, 4a, Avenida Complutense s/n, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| |
Collapse
|
44
|
β-arrestin 2 mediates the anti-inflammatory effects of fluoxetine in lipopolysaccharide-stimulated microglial cells. J Neuroimmune Pharmacol 2014; 9:582-90. [DOI: 10.1007/s11481-014-9556-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/10/2014] [Indexed: 01/28/2023]
|
45
|
Meredith GE, Rademacher DJ. MPTP mouse models of Parkinson's disease: an update. JOURNAL OF PARKINSONS DISEASE 2014; 1:19-33. [PMID: 23275799 DOI: 10.3233/jpd-2011-11023] [Citation(s) in RCA: 305] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Among the most widely used models of Parkinson's disease (PD) are those that employ toxins, especially 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Depending on the protocol used, MPTP yields large variations in nigral cell loss, striatal dopamine loss and behavioral deficits. Motor deficits do not fully replicate those seen in PD. Nonetheless, MPTP mouse models mimic many aspects of the disease and are therefore important tools for understanding PD. In this review, we will discuss the ability of MPTP mouse models to replicate the pathophysiology of PD, the mechanisms of MPTP-induced neurotoxicity, strain differences in susceptibility to MPTP, and the models' roles in testing therapeutic approaches.
Collapse
Affiliation(s)
- Gloria E Meredith
- Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, USA.
| | | |
Collapse
|
46
|
Molecular mechanisms of increased cerebral vulnerability after repeated mild blast-induced traumatic brain injury. TRANSLATIONAL PROTEOMICS 2014. [DOI: 10.1016/j.trprot.2013.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
47
|
Abstract
Microglia, the resident innate immune cells in the brain, have long been understood to be crucial to maintenance in the nervous system, by clearing debris, monitoring for infiltration of infectious agents, and mediating the brain's inflammatory and repair response to traumatic injury, stroke, or neurodegeneration. A wave of new research has shown that microglia are also active players in many basic processes in the healthy brain, including cell proliferation, synaptic connectivity, and physiology. Microglia, both in their capacity as phagocytic cells and via secretion of many neuroactive molecules, including cytokines and growth factors, play a central role in early brain development, including sexual differentiation of the brain. In this review, we present the vast roles microglia play in normal brain development and how perturbations in the normal neuroimmune environment during development may contribute to the etiology of brain-based disorders. There are notable differences between microglia and neuroimmune signaling in the male and female brain throughout the life span, and these differences may contribute to the vast differences in the incidence of neuropsychiatric and neurological disorders between males and females.
Collapse
Affiliation(s)
- Kathryn M Lenz
- Department of Psychology and Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
48
|
Mishra MK, Beaty CA, Lesniak WG, Kambhampati SP, Zhang F, Wilson MA, Blue ME, Troncoso JC, Kannan S, Johnston MV, Baumgartner WA, Kannan RM. Dendrimer brain uptake and targeted therapy for brain injury in a large animal model of hypothermic circulatory arrest. ACS NANO 2014; 8:2134-47. [PMID: 24499315 PMCID: PMC4004292 DOI: 10.1021/nn404872e] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 02/05/2014] [Indexed: 05/20/2023]
Abstract
Treatment of brain injury following circulatory arrest is a challenging health issue with no viable therapeutic options. Based on studies in a clinically relevant large animal (canine) model of hypothermic circulatory arrest (HCA)-induced brain injury, neuroinflammation and excitotoxicity have been identified as key players in mediating the brain injury after HCA. Therapy with large doses of valproic acid (VPA) showed some neuroprotection but was associated with adverse side effects. For the first time in a large animal model, we explored whether systemically administered polyamidoamine (PAMAM) dendrimers could be effective in reaching target cells in the brain and deliver therapeutics. We showed that, upon systemic administration, hydroxyl-terminated PAMAM dendrimers are taken up in the brain of injured animals and selectively localize in the injured neurons and microglia in the brain. The biodistribution in other major organs was similar to that seen in small animal models. We studied systemic dendrimer-drug combination therapy with two clinically approved drugs, N-acetyl cysteine (NAC) (attenuating neuroinflammation) and valproic acid (attenuating excitotoxicity), building on positive outcomes in a rabbit model of perinatal brain injury. We prepared and characterized dendrimer-NAC (D-NAC) and dendrimer-VPA (D-VPA) conjugates in multigram quantities. A glutathione-sensitive linker to enable for fast intracellular release. In preliminary efficacy studies, combination therapy with D-NAC and D-VPA showed promise in this large animal model, producing 24 h neurological deficit score improvements comparable to high dose combination therapy with VPA and NAC, or free VPA, but at one-tenth the dose, while significantly reducing the adverse side effects. Since adverse side effects of drugs are exaggerated in HCA, the reduced side effects with dendrimer conjugates and suggestions of neuroprotection offer promise for these nanoscale drug delivery systems.
Collapse
Affiliation(s)
- Manoj K. Mishra
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Claude A. Beaty
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Wojciech G. Lesniak
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Siva P. Kambhampati
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Fan Zhang
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Mary A. Wilson
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
| | - Mary E. Blue
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
| | - Juan C. Troncoso
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Division of Neuropathology, Department of Anesthesiology and Critical Care Medicine, and Department of Pediatrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Sujatha Kannan
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
- Division of Neuropathology, Department of Anesthesiology and Critical Care Medicine, and Department of Pediatrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Michael V. Johnston
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, Maryland 21205, United States
- Division of Neuropathology, Department of Anesthesiology and Critical Care Medicine, and Department of Pediatrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - William A. Baumgartner
- Division of Cardiac Surgery, Department of Neurology, and Department of Neuroscience, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
- Address correspondence to ,
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine/Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
- Address correspondence to ,
| |
Collapse
|
49
|
Sharma P, Ping L. Calcium ion influx in microglial cells: physiological and therapeutic significance. J Neurosci Res 2014; 92:409-23. [PMID: 24464907 DOI: 10.1002/jnr.23344] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/09/2013] [Accepted: 11/12/2013] [Indexed: 01/16/2023]
Abstract
Microglial cells, the immunocompetent cells of the central nervous system (CNS), exhibit a resting phenotype under healthy conditions. In response to injury, however, they transform into an activated state, which is a hallmark feature of many CNS diseases. Factors or agents released from the neurons, blood vessels, and/or astrocytes could activate these cells, leading to their functional and structural modifications. Microglial cells are well equipped to sense environmental changes within the brain under both physiological and pathological conditions. Entry of calcium ions (Ca(2+)) plays a critical role in the process of microglial transformation; several channels and receptors have been identified on the surface of microglial cells. These include store-operated channel, Orai1, and its sensor protein, stromal interaction molecule 1 (STIM1), in microglial cells, and their functions are modulated under pathological stimulations. Transient receptor potential (TRP) channels and voltage- and ligand-gated channels (ionotropic and metabotropic receptors) are also responsible for Ca(2+) influx into the microglial cells. An elevation of intracellular Ca(2+) concentration subsequently regulates microglial cell functions by activating a diverse array of Ca(2+)-sensitive signaling cascades. Perturbed Ca(2+) homeostasis contributes to the progression of a number of CNS disorders. Thus, regulation of Ca(2+) entry into microglial cells could be a pharmacological target for several CNS-related pathological conditions. This Review addresses the recent insights into microglial cell Ca(2+) influx mechanisms, their roles in the regulation of functions, and alterations of Ca(2+) entry in specific CNS disorders.
Collapse
Affiliation(s)
- Purnima Sharma
- All India Institute of Medical Sciences-Physiology, Basni Industrial Area Phase II Jodhpur, Rajasthan, India
| | | |
Collapse
|
50
|
Feng M, Shu Y, Yang Y, Zheng X, Li R, Wang Y, Dai Y, Qiu W, Lu Z, Hu X. Ulinastatin attenuates experimental autoimmune encephalomyelitis by enhancing anti-inflammatory responses. Neurochem Int 2013; 64:64-72. [PMID: 24274996 DOI: 10.1016/j.neuint.2013.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 11/02/2013] [Accepted: 11/07/2013] [Indexed: 10/26/2022]
Abstract
Multiple sclerosis (MS) is a common inflammatory and demyelinating neurological disease. Experimental autoimmune encephalomyelitis (EAE), an animal model of MS, has been widely used to test MS treatment methods. Ulinastatin (UTI), a drug used to treat acute inflammatory disorders, has been tested in animal models of autoimmune inflammatory diseases, such as ulcerative colitis and crescentic glomerulonephritis. We recently found that UTI has a neuroprotective effect on EAE by reducing oligodendrocyte apoptosis and demyelination. The anti-inflammatory effects of UTI on EAE/MS, however, have never been investigated. We have therefore evaluated the anti-inflammatory effects of UTI in EAE and explored the mechanisms underlying this effect. EAE was induced in mice with and without UTI treatment. Inflammation and demyelination of spinal cords were evaluated by staining with hematoxylin and eosin and with Luxol fast blue, respectively. Inflammatory markers in serum were analyzed by the Luminex method, and spinal cords were evaluated by immunofluorescence and Western blotting. UTI significantly lowered the clinical and pathological scores and the serum concentrations of the inflammatory cytokines interleukin (IL)-1β, IL-6, and matrix metal protease-9 (MMP-9). UTI also reduced the expression of tumor necrosis factor-alpha (TNF-α)/nuclear factor kappaB (NF-κB)/inducible nitric oxide synthase (iNOS) proteins and decreased CD11b(+) cells in spinal cord lesions. UTI may protect against EAE in mice by suppressing inflammatory responses. We think that UTI might be a potential therapeutic agent for MS.
Collapse
Affiliation(s)
- Ming Feng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China; Department of Gerontology and Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaqing Shu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Yu Yang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Xueping Zheng
- Department of Neurology, The Affiliated Hospital of Qingdao Medical College, Qingdao University, Qingdao 266003, China
| | - Rui Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Yongqiang Dai
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Xueqiang Hu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|