1
|
Mao Y, Meng Y, Zou K, Qin N, Wang Y, Yan J, Chen P, Cheng Y, Shi W, Zhou C, Chen H, Sheng J, Liu X, Pan J, Huang H. Advanced paternal age exacerbates neuroinflammation in offspring via m6A modification-mediated intergenerational inheritance. J Neuroinflammation 2024; 21:249. [PMID: 39367406 PMCID: PMC11453047 DOI: 10.1186/s12974-024-03248-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND The trend of postponing childbearing age is prevalent worldwide. Advanced paternal age (APA) is associated with adverse pregnancy outcomes and offspring health. However, the underlying mechanism by which paternal aging affects the risk of offspring neuropsychiatric disorders is unclear. Our study aims to explore the behavioral phenotypes and the pathologic epigenetic alterations of APA offspring inherited from aging sperm. METHODS Behavioral tests, ELISA assay, immunofluorescence and western blotting were performed on offspring mice. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA immunoprecipitation sequencing (RIP-seq) were used to investigate the modified N6-methyladenosine (m6A) profiles of paternal sperm and offspring hippocampus. Intervention of gene expression by lentivirus and adeno-associated virus in both vivo and vitro examined the potential therapeutic targets of intergenerational inherited neuroinflammation. RESULTS In our study, APA offspring exhibit cognitive impairment and autism-like behavior. An increase in neuroinflammation in APA offspring is associated with microglial overactivation, which manifests as abnormal morphology and augmented engulfment. MeRIP-seq of F0 sperm and F1 hippocampus reveal that Nr4a2 is hypermethylated with decreased expression in APA offspring involving in synaptic plasticity and microglial function. In addition, Ythdc1, an m6A reader protein, is markedly elevated in aging sperm and remains elevated in adult hippocampus of APA group. Enhanced Ythdc1 recognizes and suppresses the hypermethylated Nr4a2, thereby contributing to the abnormal phenotype in offspring. The overexpression of Ythdc1 triggers microglial activation in vitro and its suppression in the hippocampus of APA progeny alleviates behavioral aberrations and attenuates neuroinflammation. CONCLUSION Our study provides additional evidence of the abnormal behavioral phenotypes of APA offspring and reveals potential epigenetic inheritance signatures and targeted genes for future research.
Collapse
Affiliation(s)
- Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China
| | - Yicong Meng
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kexin Zou
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ningxin Qin
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinyu Wang
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Yan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - PinJia Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Weihui Shi
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Chengliang Zhou
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huixi Chen
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianzhong Sheng
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Xinmei Liu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China.
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, (No. 2019RU056), Shanghai, China.
| |
Collapse
|
2
|
Fujikawa R, Yamada J, Maeda S, Iinuma KM, Moriyama G, Jinno S. Inhibition of reactive oxygen species production accompanying alternatively activated microglia by risperidone in a mouse ketamine model of schizophrenia. J Neurochem 2024; 168:2690-2709. [PMID: 38770640 DOI: 10.1111/jnc.16133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Recent studies have highlighted the potential involvement of reactive oxygen species (ROS) and microglia, a major source of ROS, in the pathophysiology of schizophrenia. In our study, we explored how the second-generation antipsychotic risperidone (RIS) affects ROS regulation and microglial activation in the hippocampus using a mouse ketamine (KET) model of schizophrenia. KET administration resulted in schizophrenia-like behaviors in male C57BL/6J mice, such as impaired prepulse inhibition (PPI) of the acoustic startle response and hyper-locomotion. These behaviors were mitigated by RIS. We found that the gene expression level of an enzyme responsible for ROS production (Nox2), which is primarily associated with activated microglia, was lower in KET/RIS-treated mice than in KET-treated mice. Conversely, the levels of antioxidant enzymes (Ho-1 and Gclc) were higher in KET/RIS-treated mice. The microglial density in the hippocampus was increased in KET-treated mice, which was counteracted by RIS. Hierarchical cluster analysis revealed three morphological subtypes of microglia. In control mice, most microglia were resting-ramified (type I, 89.7%). KET administration shifted the microglial composition to moderately ramified (type II, 44.4%) and hyper-ramified (type III, 25.0%). In KET/RIS-treated mice, type II decreased to 32.0%, while type III increased to 34.0%. An in vitro ROS assay showed that KET increased ROS production in dissociated hippocampal microglia, and this effect was mitigated by RIS. Furthermore, we discovered that a NOX2 inhibitor could counteract KET-induced behavioral deficits. These findings suggest that pharmacological inhibition of ROS production by RIS may play a crucial role in ameliorating schizophrenia-related symptoms. Moreover, modulating microglial activation to regulate ROS production has emerged as a novel avenue for developing innovative treatments for schizophrenia.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
3
|
Diniz DG, de Oliveira JHP, Guerreiro LCF, de Menezes GC, de Assis ACL, Duarte TQ, dos Santos IBD, Maciel FD, Soares GLDS, Araújo SC, Franco FTDC, do Carmo EL, Morais RDAB, de Lima CM, Brites D, Anthony DC, Diniz JAP, Diniz CWP. Contrasting Disease Progression, Microglia Reactivity, Tolerance, and Resistance to Toxoplasma gondii Infection in Two Mouse Strains. Biomedicines 2024; 12:1420. [PMID: 39061995 PMCID: PMC11274029 DOI: 10.3390/biomedicines12071420] [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: 04/18/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 07/28/2024] Open
Abstract
Our study investigated the innate immune response to Toxoplasma gondii infection by assessing microglial phenotypic changes and sickness behavior as inflammatory response markers post-ocular tachyzoite instillation. Disease progression in Swiss albino mice was compared with the previously documented outcomes in BALB/c mice using an identical ocular route and parasite burden (2 × 105 tachyzoites), with saline as the control. Contrary to expectations, the Swiss albino mice displayed rapid, lethal disease progression, marked by pronounced sickness behaviors and mortality within 11-12 days post-infection, while the survivors exhibited no apparent signs of infection. Comparative analysis revealed the T. gondii-infected BALB/c mice exhibited reduced avoidance of feline odors, while the infected Swiss albino mice showed enhanced avoidance responses. There was an important increase in microglial cells in the dentate gyrus molecular layer of the infected Swiss albino mice compared to the BALB/c mice and their respective controls. Hierarchical cluster and discriminant analyses identified three microglial morphological clusters, differentially affected by T. gondii infection across strains. The BALB/c mice exhibited increased microglial branching and complexity, while the Swiss albino mice showed reduced shrunken microglial arbors, diminishing their morphological complexity. These findings highlight strain-specific differences in disease progression and inflammatory regulation, indicating lineage-specific mechanisms in inflammatory responses, tolerance, and resistance. Understanding these elements is critical in devising control measures for toxoplasmosis.
Collapse
Affiliation(s)
- Daniel G. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Jhonnathan H. P. de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Luma C. F. Guerreiro
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal do Pará, Campus Bragança, Bragança 68600-000, Pará, Brazil
| | - Gabriel C. de Menezes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Alexa C. L. de Assis
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Tainá Q. Duarte
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Izabelly B. D. dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Flávia D. Maciel
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Gabrielly L. da S. Soares
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Sanderson C. Araújo
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Felipe T. de C. Franco
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Ediclei L. do Carmo
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Rafaela dos A. B. Morais
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Camila M. de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Daniel C. Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - José A. P. Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Cristovam W. P. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| |
Collapse
|
4
|
Salas-Gallardo GA, Lorea-Hernández JJ, Robles-Gómez ÁA, Del Campo CCM, Peña-Ortega F. Morphological differentiation of peritumoral brain zone microglia. PLoS One 2024; 19:e0297576. [PMID: 38451958 PMCID: PMC10919594 DOI: 10.1371/journal.pone.0297576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
The Peritumoral Brain Zone (PBZ) contributes to Glioblastoma (GBM) relapse months after the resection of the original tumor, which is influenced by a variety of pathological factors. Among those, microglia are recognized as one of the main regulators of GBM progression and probably relapse. Although microglial morphology has been analyzed inside GBM and its immediate surroundings, it has not been objectively characterized throughout the PBZ. Thus, we aimed to perform a thorough characterization of microglial morphology in the PBZ and its likely differentiation not just from the tumor-associated microglia but from control tissue microglia. For this purpose, Sprague Dawley rats were intrastriatally implanted with C6 cells to induce a GBM formation. Gadolinium-based magnetic resonance imaging (MRI) was performed to locate the tumor and to define the PBZ (2 mm beyond the tumor border), thus delimitating the different regions of interest (ROIs: core tumoral zone and immediate interface; contralateral striatum as control). Brain slices were obtained and immunolabeled with the microglia marker Iba-1. Sixteen morphological parameters were measured for each cell, significative differences were found in all parameters when comparing the four ROIs. To determine if PBZ microglia could be morphologically differentiated from microglia in other ROIs, hierarchical clustering analysis was performed, revealing that microglia can be separated into four morphologically differentiated clusters, each of them mostly integrated by cells sampled in each ROI. Furthermore, a classifier based on linear discriminant analysis, including only three morphological parameters, categorized microglial cells across the studied ROIs and showed a gradual transition between them. The robustness of this classification was assessed through principal component analysis with the remaining 13 morphological parameters, corroborating the obtained results. Thus, in this study we provided objective and quantitative evidence that PBZ microglia represent a differentiable microglial morphotype that could contribute to the recurrence of GBM in this area.
Collapse
Affiliation(s)
- G. Anahí Salas-Gallardo
- Laboratorio de Células Neurales Troncales, CIACYT-Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Jonathan-Julio Lorea-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Ángel Abdiel Robles-Gómez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | - Claudia Castillo-Martin Del Campo
- Laboratorio de Células Neurales Troncales, CIACYT-Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| |
Collapse
|
5
|
Strehle LD, Otto-Dobos LD, Grant CV, Glasper ER, Pyter LM. Microglia contribute to mammary tumor-induced neuroinflammation in a female mouse model. FASEB J 2024; 38:e23419. [PMID: 38236370 PMCID: PMC10832463 DOI: 10.1096/fj.202301580rr] [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: 08/03/2023] [Revised: 11/13/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Following diagnosis but before treatment, up to 30% of breast cancer patients report behavioral side effects (e.g., anxiety, depression, memory impairment). Our rodent mammary tumor model recapitulates aspects of these behavioral sequelae, as well as elevated circulating and brain inflammatory mediators. Neuroinflammation is a proposed mechanism underlying the etiology of mood disorders and cognitive deficits, and therefore may be contributing to tumor-associated behavioral side effects. The cellular mechanisms by which tumor-induced neuroinflammation occurs remain unknown, making targeted treatment approaches inaccessible. Here, we tested the hypotheses that microglia are the primary cells driving tumor-induced neuroinflammation and behavioral side effects. Young adult female BALB/c mice were induced with a 67NR mammary tumor; tumor-free controls underwent a sham surgery. Mammary tumors increased IBA1+ and GFAP+ staining in the amygdala and hippocampus relative to tumor-free controls. However, tumors did not alter gene expression of Percoll-enriched microglia isolated from the whole brain. While cognitive, social, and anhedonia-like behaviors were not altered in tumor-bearing mice, tumors increased central tendency in the open-field test; microglia depletion did not reverse this effect. Brain region RT-qPCR data indicated that microglia depletion attenuated tumor-induced elevations of neuroinflammatory gene expression in a region- and mediator-specific manner. These results indicate a causal role of microglia in tumor-induced neuroinflammation. This research advances our understanding of the cellular mechanisms underlying tumor-induced neuroinflammation in order to understand how brain responses (e.g., behavior) may be altered with subsequent cancer-related immune challenges.
Collapse
Affiliation(s)
- Lindsay D. Strehle
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lauren D. Otto-Dobos
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Corena V. Grant
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Erica R. Glasper
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Leah M. Pyter
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
Fujikawa R, Jinno S. Identification of hyper-ramified microglia in the CA1 region of the mouse hippocampus potentially associated with stress resilience. Eur J Neurosci 2022; 56:5137-5153. [PMID: 36017697 DOI: 10.1111/ejn.15812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 12/14/2022]
Abstract
Recent studies have indicated that some individuals are less affected by stress, and such individuals are called resilient. This study aimed to determine whether the specific phenotype of microglia might be involved in resilience using the social defeat stress paradigm. Male C57BL/6J (B6) mice were attacked by aggressive male ICR mice for five consecutive days. After stress exposure, the social behaviour was reduced in about half of the B6 mice (vulnerable), whereas no such change was observed in the remaining half of the B6 mice (resilient). Anxiety-like behaviour was increased in vulnerable mice compared with resilient mice and non-stressed controls. However, depression-related behaviour was comparable between the three groups. The morphological characteristics of microglia in the CA1 region of the dorsal hippocampus in non-stressed controls and resilient mice differed from those in vulnerable mice. Interestingly, the voxel densities of GABAergic and glutamatergic synaptic puncta colocalized with microglia were higher in resilient mice than in non-stressed controls and vulnerable mice. Microglia were then objectively classified into three morphological types by hierarchical cluster analysis. The appearance of type I microglia resembled the so-called resting ramified microglia and represented the major population of microglia in non-stressed controls. Type II microglia exhibited a de-ramified morphology and accounted for 60% of the microglia in vulnerable mice. Type III microglia showed a hyper-ramified morphology and represented more than half of the microglia in resilient mice. These results suggest that hyper-ramified microglia in the hippocampus may be associated with stress resilience via the modulation of synaptic transmission.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
7
|
Gong L, Zhu T, Chen C, Xia N, Yao Y, Ding J, Xu P, Li S, Sun Z, Dong X, Shen W, Sun P, Zeng L, Xie Y, Jiang P. Miconazole exerts disease-modifying effects during epilepsy by suppressing neuroinflammation via NF-κB pathway and iNOS production. Neurobiol Dis 2022; 172:105823. [PMID: 35878745 DOI: 10.1016/j.nbd.2022.105823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 11/29/2022] Open
Abstract
Neuroinflammation contributes to the generation of epilepsy and has been proposed as an effective therapeutic target. Recent studies have uncovered the potential effects of the anti-fungal drug miconazole for treating various brain diseases by suppressing neuroinflammation but have not yet been studied in epilepsy. Here, we investigated the effects of different doses of miconazole (5, 20, 80 mg/kg) on seizure threshold, inflammatory cytokines release, and glial cells activation in the pilocarpine (PILO) pentylenetetrazole (PTZ), and intrahippocampal kainic acid (IHKA) models. We demonstrated that 5 and 20 mg/kg miconazole increased seizure threshold, but only 20 mg/kg miconazole reduced inflammatory cytokines release, glial cells activation, and morphological alteration during the early post-induction period (24 h, 3 days). We further investigated the effects of 20 mg/kg miconazole on epilepsy (4 weeks after KA injection). We found that miconazole significantly attenuated cytokines production, glial cells activation, microglial morphological changes, frequency and duration of recurrent hippocampal paroxysmal discharges (HPDs), and neuronal and synaptic damage in the hippocampus during epilepsy. In addition, miconazole suppressed the KA-induced activation of the NF-κB pathway and iNOS production. Our results indicated miconazole to be an effective drug for disease-modifying effects during epilepsy, which may act by attenuating neuroinflammation through the suppression of NF-κB activation and iNOS production. At appropriate doses, miconazole may be a safe and effective approved drug that can easily be repositioned for clinical practice.
Collapse
Affiliation(s)
- Lifen Gong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China
| | - Chen Chen
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Ningxiao Xia
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Yinping Yao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Shaoxing People's Hospital, Shaoxing 312300, China
| | - Junchao Ding
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Yiwu Maternity and Children Hospital, Yiwu 322000, China
| | - Peng Xu
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Tongxiang First People's Hospital, Tongxiang 314500, China
| | - Shufen Li
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Lishui Center Hospital, Lishui 323000, China
| | - Zengxian Sun
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Lishui Center Hospital, Lishui 323000, China
| | - Xinyan Dong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Weida Shen
- Department of Pharmacy, Zhejiang University City College School of Medicine, Hangzhou 310015, China
| | - Peng Sun
- Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Linghui Zeng
- Department of Pharmacy, Zhejiang University City College School of Medicine, Hangzhou 310015, China.
| | - Yicheng Xie
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China.
| | - Peifang Jiang
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China.
| |
Collapse
|
8
|
de Almeida Miranda D, Araripe J, de Morais Magalhães NG, de Siqueira LS, de Abreu CC, Pereira PDC, Henrique EP, da Silva Chira PAC, de Melo MAD, do Rêgo PS, Diniz DG, Sherry DF, Diniz CWP, Guerreiro-Diniz C. Shorebirds' Longer Migratory Distances Are Associated With Larger ADCYAP1 Microsatellites and Greater Morphological Complexity of Hippocampal Astrocytes. Front Psychol 2022; 12:784372. [PMID: 35185684 PMCID: PMC8855117 DOI: 10.3389/fpsyg.2021.784372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
For the epic journey of autumn migration, long-distance migratory birds use innate and learned information and follow strict schedules imposed by genetic and epigenetic mechanisms, the details of which remain largely unknown. In addition, bird migration requires integrated action of different multisensory systems for learning and memory, and the hippocampus appears to be the integration center for this task. In previous studies we found that contrasting long-distance migratory flights differentially affected the morphological complexity of two types of hippocampus astrocytes. Recently, a significant association was found between the latitude of the reproductive site and the size of the ADCYAP1 allele in long distance migratory birds. We tested for correlations between astrocyte morphological complexity, migratory distances, and size of the ADCYAP1 allele in three long-distance migrant species of shorebird and one non-migrant. Significant differences among species were found in the number and morphological complexity of the astrocytes, as well as in the size of the microsatellites of the ADCYAP1 gene. We found significant associations between the size of the ADCYAP1 microsatellites, the migratory distances, and the degree of morphological complexity of the astrocytes. We suggest that associations between astrocyte number and morphological complexity, ADCYAP1 microsatellite size, and migratory behavior may be part of the adaptive response to the migratory process of shorebirds.
Collapse
Affiliation(s)
- Diego de Almeida Miranda
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil.,Laboratório de Genética e Conservação, Instituto de Estudos Costeiros (IECOS), Universidade Federal do Pará, Bragança, Brazil
| | - Juliana Araripe
- Laboratório de Genética e Conservação, Instituto de Estudos Costeiros (IECOS), Universidade Federal do Pará, Bragança, Brazil
| | - Nara G de Morais Magalhães
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Lucas Silva de Siqueira
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Cintya Castro de Abreu
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Patrick Douglas Corrêa Pereira
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Ediely Pereira Henrique
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Pedro Arthur Campos da Silva Chira
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Mauro A D de Melo
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Péricles Sena do Rêgo
- Laboratório de Genética e Conservação, Instituto de Estudos Costeiros (IECOS), Universidade Federal do Pará, Bragança, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Belém, Brazil.,Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Brazil
| | - David Francis Sherry
- Department of Psychology, Advanced Facility for Avian Research, University of Western Ontario, London, ON, Canada
| | - Cristovam W P Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Cristovam Guerreiro-Diniz
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| |
Collapse
|
9
|
The Effects of Modified Curcumin Preparations on Glial Morphology in Aging and Neuroinflammation. Neurochem Res 2022; 47:813-824. [PMID: 34988899 DOI: 10.1007/s11064-021-03499-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is characterized by reactive microglia and astrocytes (collectively called gliosis) in the central nervous system and is considered as one of the main pathological hallmarks in different neurodegenerative diseases such as Alzheimer's disease, age-related dementia, and multiple sclerosis. Upon activation, glia undergoes structural and morphological changes such as the microglial cells swell in size and astrocytes become bushy, which play both beneficial and detrimental roles. Hence, they are unable to perform the normal physiological role in brain immunity. Curcumin, a cytokine suppressive anti-inflammatory drug, has a high proven pre-clinical potency and efficacy to reverse chronic neuroinflammation by attenuating the activation and morphological changes that occur in the microglia and astrocytes. This review will highlight the recent findings on the tree structure changes of microglia and astrocytes in neuroinflammation and the effects of curcumin against the activation and morphology of glial cells.
Collapse
|
10
|
Phytoestrogen genistein modulates neuron-microglia signaling in a mouse model of chronic social defeat stress. Neuropharmacology 2022; 206:108941. [PMID: 34990615 DOI: 10.1016/j.neuropharm.2021.108941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 01/20/2023]
Abstract
Microglia, resident immune cells in the brain, are shown to mediate the crosstalk between psychological stress and depression. Interestingly, increasing evidence indicates that sex hormones, particularly estrogen, are involved in the regulation of immune system. In this study, we aimed to understand the potential effects of chronic social defeat stress (CSDS) and genistein (GEN), an estrogenic compound of the plant origin, on neuron-microglia interactions in the mouse hippocampus. The time spent in the avoidance zone in the social interaction test was increased by CSDS 1 day after the exposure, while the avoidance behavior returned to control levels 14 days after the CSDS exposure. Similar results were obtained from the elevated plus-maze test. However, the immobility time in the forced swim test was increased by CSDS 14 days after the exposure, and the depression-related behavior was in part alleviated by GEN. The numerical densities of microglia in the hippocampus were increased by CSDS, and they were decreased by GEN. The voxel densities of synaptic structures and synaptic puncta colocalized with microglia were decreased by CSDS, and they were increased by GEN. Neither CSDS nor GEN affected the gene expressions of major pro-inflammatory cytokines. Conversely, the expression levels of genes related to neurotrophic factors were decreased by CSDS, and they were partially reversed by GEN. These findings show that GEN may in part alleviate stress-related symptoms, and the effects of GEN may be associated with the modulation of neuron-microglia signaling via chemokines and neurotrophic factors in the hippocampus.
Collapse
|
11
|
Role of neuroglia in neuropathic pain and depression. Pharmacol Res 2021; 174:105957. [PMID: 34688904 DOI: 10.1016/j.phrs.2021.105957] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/06/2021] [Accepted: 10/20/2021] [Indexed: 12/30/2022]
Abstract
Patients with neuropathic pain induced by nerve injury usually present with co-morbid affective changes, such as depression. Neuroglia was reported to play an important role in the development and maintenance of neuropathic pain both centrally and peripherally. Meanwhile, there have been studies showing that neuroglia participated in the development of depression. However, the specific role of neuroglia in neuropathic pain and depression has not been reviewed comprehensively. Therefore, we summarized the recent findings on the role of neuroglia in neuropathic pain and depression. Based on this review, we found a bridge-like role of neuroglia in neuropathic pain co-morbid with depression. This review may provide therapeutic implications in the treatment of neuropathic pain and offer potential help in the studies of mechanisms in the future.
Collapse
|
12
|
da Silva Creão LS, Neto JBT, de Lima CM, dos Reis RR, de Sousa AA, dos Santos ZA, Diniz JAP, Diniz DG, Diniz CWP. Microglial Metamorphosis in Three Dimensions in Virus Limbic Encephalitis: An Unbiased Pictorial Representation Based on a Stereological Sampling Approach of Surveillant and Reactive Microglia. Brain Sci 2021; 11:brainsci11081009. [PMID: 34439628 PMCID: PMC8393838 DOI: 10.3390/brainsci11081009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 12/03/2022] Open
Abstract
Microglia influence pathological progression in neurological diseases, reacting to insults by expressing multiple morphofunctional phenotypes. However, the complete morphological spectrum of reactive microglia, as revealed by three-dimensional microscopic reconstruction, has not been detailed in virus limbic encephalitis. Here, using an anatomical series of brain sections, we expanded on an earlier Piry arbovirus encephalitis study to include CA1/CA2 and assessed the morphological response of homeostatic and reactive microglia at eight days post-infection. Hierarchical cluster and linear discriminant function analyses of multimodal morphometric features distinguished microglial morphology between infected animals and controls. For a broad representation of the spectrum of microglial morphology in each defined cluster, we chose representative cells of homeostatic and reactive microglia, using the sum of the distances of each cell in relation to all the others. Based on multivariate analysis, reactive microglia of infected animals showed more complex trees and thicker branches, covering a larger volume of tissue than in control animals. This approach offers a reliable representation of microglia dispersion in the Euclidean space, revealing the morphological kaleidoscope of surveillant and reactive microglia morphotypes. Because form precedes function in nature, our findings offer a starting point for research using integrative methods to understand microglia form and function.
Collapse
Affiliation(s)
- Leonardo Sávio da Silva Creão
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Brazil; (L.S.d.S.C.); (C.W.P.D.)
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - João Bento Torres Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
- Faculdade de Fisioterapia e Terapia Ocupacional, Universidade Federal do Pará, Belém 66075-110, Brazil
| | - Camila Mendes de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - Renata Rodrigues dos Reis
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - Aline Andrade de Sousa
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | - Zaire Alves dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| | | | - Daniel Guerreiro Diniz
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Brazil; (L.S.d.S.C.); (C.W.P.D.)
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66093-020, Brazil;
- Correspondence:
| | - Cristovam Wanderley Picanço Diniz
- Núcleo de Pesquisas em Oncologia, Programa de Pós-Graduação em Oncologia e Ciências Médicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Brazil; (L.S.d.S.C.); (C.W.P.D.)
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém 66073-005, Brazil; (J.B.T.N.); (C.M.d.L.); (R.R.d.R.); (A.A.d.S.); (Z.A.d.S.)
| |
Collapse
|
13
|
Li Y, Tang W, Guo M. The Cell as Matter: Connecting Molecular Biology to Cellular Functions. MATTER 2021; 4:1863-1891. [PMID: 35495565 PMCID: PMC9053450 DOI: 10.1016/j.matt.2021.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Viewing cell as matter to understand the intracellular biomolecular processes and multicellular tissue behavior represents an emerging research area at the interface of physics and biology. Cellular material displays various physical and mechanical properties, which can strongly affect both intracellular and multicellular biological events. This review provides a summary of how cells, as matter, connect molecular biology to cellular and multicellular scale functions. As an impact in molecular biology, we review recent progresses in utilizing cellular material properties to direct cell fate decisions in the communities of immune cells, neurons, stem cells, and cancer cells. Finally, we provide an outlook on how to integrate cellular material properties in developing biophysical methods for engineered living systems, regenerative medicine, and disease treatments.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wenhui Tang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
14
|
Campos RM, Barbosa-Silva MC, Ribeiro-Resende VT. Comparison of effect of crush or transection peripheral nerve lesion on lumbar spinal cord synaptic plasticity and microglial dynamics. IBRO Neurosci Rep 2021; 10:225-235. [PMID: 34179871 PMCID: PMC8211924 DOI: 10.1016/j.ibneur.2021.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022] Open
Abstract
In an injury to the peripheral nervous system, the spinal cord and brain structure reorganize connections to optimize the function of the remaining parts. Many cell events are triggered in the spinal cord to support changes in the synaptic connections around motoneurons, where old connections are removed, and new ones created. Microglial cells are primitive macrophages that invade the central nervous system in early stages of neurodevelopment and have several functions, such as eliminating synapses. We investigated the synaptic plasticity after different types of peripheral (sciatic) nerve injury (crush or total transection), as well as the behavior of microglial cells for 2 weeks after a peripheral lesion. As expected, sciatic-nerve injury reduced motor performance in mice, but crushed animals regained partial motor control. Because of sciatic-nerve injury, pre-synaptic inputs decreased around the motoneurons in the ventro-lateral horn, while microglial cells increased around these cells. Microglial cells also exhibited altered morphology in both types of peripheral lesion, indicating a similar underlying mechanism of plasticity. To investigate the involvement of microglia in this scenario, microglial activation was modulated by daily administration of minocycline. The minocycline treatment directly affected the microglial response and impacted the synapse rearrangement in the spinal cord. Together, these results demonstrate that microglia cells are involved in synaptic plasticity in the lumbar spinal cord in both nerve-injury scenarios. SUMMARY OF STATEMENT Here, we demonstrated that acute plasticity in the lumbar spinal cord (LSC) did not differ between crush and transection of peripheral nerve, and that microglial reactivity in the LSC was important after both injury types.
Collapse
Affiliation(s)
- Raquel M.P. Campos
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Neuroquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Maria Carolina Barbosa-Silva
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Neuroquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Victor T. Ribeiro-Resende
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Neuroquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ 25255-030, Brazil
| |
Collapse
|
15
|
Tanaka T, Hirai S, Hosokawa M, Saito T, Sakuma H, Saido T, Hasegawa M, Okado H. Early-life stress induces the development of Alzheimer's disease pathology via angiopathy. Exp Neurol 2021; 337:113552. [PMID: 33309748 DOI: 10.1016/j.expneurol.2020.113552] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/25/2020] [Accepted: 12/06/2020] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is a major societal, scientific, and economic problem. Several early-life factors associated with an increased risk for the clinical diagnosis of AD have recently been identified. In the present study, we investigated the involvement of early-life stress in the pathogenesis of AD using heterozygous amyloid precursor protein (APP) mutant mice (AppNL-G-F/wt) and wild-type (Appwt/wt) mice. We found that maternal-separated Appwt/wt mice showed narrowing of vessels and decreased pericyte coverage of capillaries in the prefrontal cortex, while maternal-separated AppNL-G-F/wt mice additionally showed the impairment of cognitive function, earlier formation of Aβ plaques, increased vessel-associated microglia, and disruption of the blood-brain barrier. Substantial activation of microglia was detected in the maternal-separated AppNL-G-F/wt mice and maternal-separated Appwt/wt mice. At an early stage, morphological changes and inflammatory responses were observed in the microglia of the maternal-separated AppNL-G-F/wt mice and maternal-separated Appwt/wt mice, and morphological changes in the microglia were observed in the non-maternal-separated AppNL-G-F/wt mice. Microglia activation induced by maternal separation in combination with the APP mutation may impair the vascular system, leading to AD progression. These findings therefore suggest that maternal separation results in the early induction of AD-related pathology via angiopathy.
Collapse
Affiliation(s)
- Tomoko Tanaka
- Laboratory of Neural Development, Department of Psychiatry & Behavioral Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shinobu Hirai
- Laboratory of Neural Development, Department of Psychiatry & Behavioral Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hosokawa
- Dementia Research Project, Department of Brain & Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Hiroshi Sakuma
- Child brain Project, Department of Brain & Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Masato Hasegawa
- Dementia Research Project, Department of Brain & Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Haruo Okado
- Laboratory of Neural Development, Department of Psychiatry & Behavioral Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| |
Collapse
|
16
|
Chu X, Wang C, Wu Z, Fan L, Tao C, Lin J, Chen S, Lin Y, Ge Y. JNK/c-Jun-driven NLRP3 inflammasome activation in microglia contributed to retinal ganglion cells degeneration induced by indirect traumatic optic neuropathy. Exp Eye Res 2020; 202:108335. [PMID: 33141050 DOI: 10.1016/j.exer.2020.108335] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Indirect traumatic optic neuropathy (ITON) is a major cause of permanent loss of vision after blunt head trauma. Neuroinflammation plays a crucial role in neurodegenerative diseases. The present study concentrated on JNK/c-Jun-driven NLRP3 inflammasome activation in microglia during the degeneration of retinal ganglion cells (RGCs) in ITON. METHODS An impact acceleration (IA) model was employed to induce ITON, which could produce significant neurodegeneration in the visual system. Pharmacological approaches were employed to disrupt JNK and to explore whether JNK and the microglial response contribute to RGC death and axonal degeneration. RESULTS Our results indicated that the ITON model induced significant RGC death and axonal degeneration and activated JNK/c-Jun signaling, which could further induce the microglial response and NLRP3 inflammasome activation. Moreover, JNK disruption is sufficient to suppress NLRP3 inflammasome activation in microglia and to prevent RGC death and axonal degeneration. CONCLUSIONS ITON could promote JNK/c-Jun signaling, which further activates the NLRP3 inflammasome in microglia and contributes to the degeneration of axons and death of RGCs. JNK inhibition is able to suppress the inflammatory reaction and improve RGC survival. Although further work is needed to determine whether pharmacological inhibition of the NLRP3 inflammasome can prevent ITON, our findings indicated that such intervention could be promising for translational work.
Collapse
Affiliation(s)
- Xiaoqi Chu
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Chun Wang
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Zheng Wu
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Liting Fan
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Chunmei Tao
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Jiaqi Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Shuang Chen
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China
| | - Yongzhong Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China.
| | - Yusong Ge
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian City, Liaoning Province, 116023, China.
| |
Collapse
|
17
|
Henrique EP, Oliveira MA, Paulo DC, Pereira PDC, Dias C, Siqueira LS, Lima CM, Miranda DDA, Rego PS, Araripe J, Melo MAD, Diniz DG, Morais Magalhães NG, Sherry DF, Picanço Diniz CW, Diniz CG. Contrasting migratory journeys and changes in hippocampal astrocyte morphology in shorebirds. Eur J Neurosci 2020; 54:5687-5704. [DOI: 10.1111/ejn.14781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 04/26/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Ediely Pereira Henrique
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| | - Marcus Augusto Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto Instituto de Ciências Biológicas Universidade Federal do Pará Belém Pará Brazil
| | - Dario Carvalho Paulo
- Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto Instituto de Ciências Biológicas Universidade Federal do Pará Belém Pará Brazil
| | - Patrick Douglas Corrêa Pereira
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| | - Cleyssian Dias
- Curso de Pós‐Graduação em Zoologia Museu Paraense Emílio Goeldi Universidade Federal do Pará Belém Pará Brazil
| | - Lucas Silva Siqueira
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| | - Camila Mendes Lima
- Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto Instituto de Ciências Biológicas Universidade Federal do Pará Belém Pará Brazil
| | - Diego de Almeida Miranda
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| | - Péricles Sena Rego
- Instituto de Estudos Costeiros Universidade Federal do Pará Bragança Pará Brazil
| | - Juliana Araripe
- Instituto de Estudos Costeiros Universidade Federal do Pará Bragança Pará Brazil
| | - Mauro André Damasceno Melo
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto Instituto de Ciências Biológicas Universidade Federal do Pará Belém Pará Brazil
- Instituto Evandro Chagas Laboratório de Miscroscopia Eletrônica Belém Pará Brazil
| | - Nara Gyzely Morais Magalhães
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| | - David Francis Sherry
- Department of Psychology Advanced Facility for Avian Research University of Western Ontario London ON Canada
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto Instituto de Ciências Biológicas Universidade Federal do Pará Belém Pará Brazil
| | - Cristovam Guerreiro Diniz
- Laboratório de Biologia Molecular e Neuroecologia Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança Bragança Pará Brazil
| |
Collapse
|
18
|
A Clonal NG2-Glia Cell Response in a Mouse Model of Multiple Sclerosis. Cells 2020; 9:cells9051279. [PMID: 32455842 PMCID: PMC7291195 DOI: 10.3390/cells9051279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 02/01/2023] Open
Abstract
NG2-glia, also known as oligodendrocyte precursor cells (OPCs), have the potential to generate new mature oligodendrocytes and thus, to contribute to tissue repair in demyelinating diseases like multiple sclerosis (MS). Once activated in response to brain damage, NG2-glial cells proliferate, and they acquire a reactive phenotype and a heterogeneous appearance. Here, we set out to investigate the distribution and phenotypic diversity of NG2-glia relative to their ontogenic origin, and whether there is a clonal NG2-glial response to lesion in an experimental autoimmune encephalomyelitis (EAE) murine model of MS. As such, we performed in utero electroporation of the genomic lineage tracer, StarTrack, to follow the fate of NG2-glia derived from single progenitors and to evaluate their response to brain damage after EAE induction. We then analyzed the dispersion of the NG2-glia derived clonally from single pallial progenitors in the brain of EAE mice. In addition, we examined several morphological parameters to assess the degree of NG2-glia reactivity in clonally-related cells. Our results reveal the heterogeneity of these progenitors and their cell progeny in a scenario of autoimmune demyelination, revealing the ontogenic phenomena at play in these processes.
Collapse
|
19
|
Honig MG, Dorian CC, Worthen JD, Micetich AC, Mulder IA, Sanchez KB, Pierce WF, Del Mar NA, Reiner A. Progressive long-term spatial memory loss following repeat concussive and subconcussive brain injury in mice, associated with dorsal hippocampal neuron loss, microglial phenotype shift, and vascular abnormalities. Eur J Neurosci 2020; 54:5844-5879. [PMID: 32090401 PMCID: PMC7483557 DOI: 10.1111/ejn.14711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
There is considerable concern about the long‐term deleterious effects of repeat head trauma on cognition, but little is known about underlying mechanisms and pathology. To examine this, we delivered four air blasts to the left side of the mouse cranium, a week apart, with an intensity that causes deficits when delivered singly and considered “concussive,” or an intensity that does not yield significant deficits when delivered singly and considered “subconcussive.” Neither repeat concussive nor subconcussive blast produced spatial memory deficits at 4 months, but both yielded deficits at 14 months, and dorsal hippocampal neuron loss. Hierarchical cluster analysis of dorsal hippocampal microglia across the three groups based on morphology and expression of MHCII, CX3CR1, CD68 and IBA1 revealed five distinct phenotypes. Types 1A and 1B microglia were more common in sham mice, linked to better neuron survival and memory, and appeared mildly activated. By contrast, 2B and 2C microglia were more common in repeat concussive and subconcussive mice, linked to poorer neuron survival and memory, and characterized by low expression levels and attenuated processes, suggesting they were de‐activated and dysfunctional. In addition, endothelial cells in repeat concussive mice exhibited reduced CD31 and eNOS expression, which was correlated with the prevalence of type 2B and 2C microglia. Our findings suggest that both repeat concussive and subconcussive head injury engender progressive pathogenic processes, possibly through sustained effects on microglia that over time lead to increased prevalence of dysfunctional microglia, adversely affecting neurons and blood vessels, and thereby driving neurodegeneration and memory decline.
Collapse
Affiliation(s)
- Marcia G Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Conor C Dorian
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - John D Worthen
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anthony C Micetich
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Isabelle A Mulder
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Katelyn B Sanchez
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - William F Pierce
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel A Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
20
|
Wei J, Wang M, Jing C, Keep RF, Hua Y, Xi G. Multinucleated Giant Cells in Experimental Intracerebral Hemorrhage. Transl Stroke Res 2020; 11:1095-1102. [PMID: 32090277 DOI: 10.1007/s12975-020-00790-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/16/2023]
Abstract
Macrophage phagocytosis plays an important role in hematoma clearance after intracerebral hemorrhage (ICH). This study examined the characteristics of multinucleated giant cells (MGCs), a group of macrophages with multiple nuclei, in a mouse ICH model. Whether MGCs could be increased by treatment with a CD47 blocking antibody and decreased by treatment with clodronate liposomes were also examined. ICH was induced via autologous blood injection. Male adult C57BL/6 mice in different groups had (1) ICH alone; (2) ICH with anti-CD47 blocking antibody or control IgG; and (3) ICH with anti-CD47 antibody combined with clodronate liposomes or control liposomes. The effect of anti-CD47 antibody on MGC formation was also tested in females. Brains were harvested at days 3 or 7 for brain histology. Many MGCs were found at day 3 post-ICH, but were reduced at day 7. MGCs phagocytosed many red blood cells and were heme oxygenase-1, ferritin, YM-1, and iNOS positive. CD47 blocking antibody injection increased MGC numbers in the peri-hematomal zone and in the hematoma in both sexes. Co-injection of clodronate liposomes depleted MGCs in both the hematoma core and the peri-hematomal area. In conclusion, MGCs represent a macrophage/microglia subtype with strong phagocytosis capacity. MGCs exhibited not only an M2 but also an M1 phenotype and appeared involved in hemoglobin degradation. Anti-CD47 antibody boosted the number of MGCs, which may contribute to enhance hematoma clearance. Understanding the exact roles of MGCs in ICH may reveal novel targets for ICH treatment.
Collapse
Affiliation(s)
- Jialiang Wei
- Department of Neurosurgery, R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.,Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ming Wang
- Department of Neurosurgery, R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Chaohui Jing
- Department of Neurosurgery, R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, R5018 BSRB, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
21
|
García-Magro N, Martin YB, Palomino-Antolin A, Egea J, Negredo P, Avendaño C. Multiple Morphometric Assessment of Microglial Cells in Deafferented Spinal Trigeminal Nucleus. Front Neuroanat 2020; 13:103. [PMID: 32038181 PMCID: PMC6987390 DOI: 10.3389/fnana.2019.00103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Microglia (MG) are the first cells to react to the abnormal incoming signals that follow an injury of sensory nerves and play a critical role in the development and maintenance of neuropathic pain, a common sequel of nerve injuries. Here we present population data on cell number, soma size, and length of processes of MG in the caudal division of the spinal trigeminal nucleus (Sp5C) in control mice and at the peak of microgliosis (7 days) following unilateral transection of the infraorbital nerve (IoN). The study is performed combining several bias- and assumption-free imaging and stereological approaches with different immunolabeling procedures, with the objective of tackling some hard problems that often hinder proper execution of MG morphometric studies. Our approach may easily be applied to low-density MG populations, but also works, with limited biases, in territories where MG cell bodies and processes form dense meshworks. In controls, and contralaterally to the deafferented side, MG cell body size and shape and branching pattern matched well the descriptions of “resting” or “surveillant” MG described elsewhere, with only moderate intersubject variability. On the superficial laminae of the deafferented side, however, MG displayed on average larger somata and remarkable diversity in shape. The number of cells and the length of MG processes per mm3 increased 5 and 2.5 times, respectively, indicating a net 50% decrease in the mean length of processes per cell. By using specific immunolabeling and cell sorting of vascular macrophages, we found only a negligible fraction of these cells in Sp5C, with no differences between controls and deafferented animals, suggesting that blood-borne monocytes play at most a very limited role in the microgliosis occurring following sensory nerve deafferentation. In sum, here we present reliable morphometric data on MG in control and deafferented trigeminal nuclei using efficient methods that we propose may equally be applied to any morphometric population analysis of these cells under different physiological or pathological conditions.
Collapse
Affiliation(s)
- Nuria García-Magro
- Department of Anatomy, Histology and Neuroscience, Medical School, Autonomous University of Madrid, Madrid, Spain.,Ph.D. Programme in Neuroscience, Doctoral School, Autonomous University of Madrid, Madrid, Spain
| | - Yasmina B Martin
- Department of Anatomy, Faculty of Medicine, Francisco de Vitoria University, Madrid, Spain
| | - Alejandra Palomino-Antolin
- Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| | - Javier Egea
- Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| | - Pilar Negredo
- Department of Anatomy, Histology and Neuroscience, Medical School, Autonomous University of Madrid, Madrid, Spain
| | - Carlos Avendaño
- Department of Anatomy, Histology and Neuroscience, Medical School, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
22
|
Distinct P2Y Receptors Mediate Extension and Retraction of Microglial Processes in Epileptic and Peritumoral Human Tissue. J Neurosci 2020; 40:1373-1388. [PMID: 31896671 DOI: 10.1523/jneurosci.0218-19.2019] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.
Collapse
|
23
|
Fernández-Arjona MDM, Grondona JM, Fernández-Llebrez P, López-Ávalos MD. Microglial Morphometric Parameters Correlate With the Expression Level of IL-1β, and Allow Identifying Different Activated Morphotypes. Front Cell Neurosci 2019; 13:472. [PMID: 31708746 PMCID: PMC6824358 DOI: 10.3389/fncel.2019.00472] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022] Open
Abstract
Microglia are the resident macrophages in the brain. Traditionally, two forms of microglia have been described: one considered as a resting/surveillant state in which cells have a highly branched morphology, and another considered as an activated state in which they acquire a de-ramified or amoeboid form. However, many studies describe intermediate microglial morphologies which emerge during pathological processes. Since microglial form and function are closely related, it is of interest to correlate microglial morphology with the extent of its activation. To address this issue, we used a rat model of neuroinflammation consisting in a single injection of the enzyme neuraminidase (NA) within the lateral ventricle. Sections from NA-injected animals were co-immunolabeled with the microglial marker IBA1 and the cytokine IL-1β, which highlight features of the cell’s shape and inflammatory activation, respectively. Activated (IL-1β positive) microglial cells were sampled from the dorsal hypothalamus nearby the third ventricle. Images of single microglial cells were processed in two different ways to obtain (1) an accurate measure of the level of expression of IL-1β (indicating the degree of activation), and (2) a set of 15 morphological parameters to quantitatively and objectively describe the cell’s shape. A simple regression analysis revealed a dependence of most of the morphometric parameters on IL-1β expression, demonstrating that the morphology of microglial cells changes progressively with the degree of activation. Moreover, a hierarchical cluster analysis pointed out four different morphotypes of activated microglia, which are characterized not only by morphological parameters values, but also by specific IL-1β expression levels. Thus, these results demonstrate in an objective manner that the activation of microglial cells is a gradual process, and correlates with their morphological change. Even so, it is still possible to categorize activated cells according to their morphometric parameters, each category presenting a different activation degree. The physiological relevance of those activated morphotypes is an issue worth to be assessed in the future.
Collapse
Affiliation(s)
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Pedro Fernández-Llebrez
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| |
Collapse
|
24
|
Fiore NT, Austin PJ. Peripheral Nerve Injury Triggers Neuroinflammation in the Medial Prefrontal Cortex and Ventral Hippocampus in a Subgroup of Rats with Coincident Affective Behavioural Changes. Neuroscience 2019; 416:147-167. [DOI: 10.1016/j.neuroscience.2019.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/25/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022]
|
25
|
Shtaya A, Bridges LR, Esiri MM, Lam‐Wong J, Nicoll JAR, Boche D, Hainsworth AH. Rapid neuroinflammatory changes in human acute intracerebral hemorrhage. Ann Clin Transl Neurol 2019; 6:1465-1479. [PMID: 31402627 PMCID: PMC6689697 DOI: 10.1002/acn3.50842] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Spontaneous intracerebral hemorrhage (ICH) is the commonest form of hemorrhagic stroke and is associated with a poor prognosis. Neurosurgical removal of intracerebral hematoma has limited benefit and no pharmacotherapies are available. In acute ICH, primary tissue damage is followed by secondary pathology, where the cellular and neuroinflammatory changes are poorly understood. METHODS We studied histological changes in postmortem tissue from a cohort of spontaneous supra-tentorial primary ICH cases (n = 27) with survival of 1-12 days, compared to a matched control group (n = 16) examined in corresponding regions. Hematoxylin-eosin and microglial (Iba1) immunolabelled sections were assessed at 0-2, 3-5, and 7-12 days post-ICH. RESULTS Peri-hematoma, the observed ICH-related changes include edema, tissue neutrophils and macrophages from day 1. Ischemic neurons and swollen endothelial cells were common at day 1 and universal after day 5, as were intramural erythrocytes within small vessel walls. Activated microglia were evident at day 1 post-ICH. There was a significant increase in Iba1 positive area fraction at 0-2 (threefold), 3-5 (fourfold), and 7-12 days post ICH (ninefold) relative to controls. Giant microglia were detected peri-hematoma from day 5 and consistently 7-12 days post-ICH. INTERPRETATION Our data indicate that neuroinflammatory processes commence from day 1 post-ICH with changing microglial size and morphology following ICH and up to day 12. From day 5 some microglia exhibit a novel multiply nucleated morphology, which may be related to changing phagocytic function. Understanding the time course of neuroinflammatory changes, post-ICH may reveal novel targets for therapy and brain restoration.
Collapse
Affiliation(s)
- Anan Shtaya
- Neuroscience Research Centre, Molecular and Clinical Sciences Research InstituteSt. George’s, University of LondonLondonUK
| | - Leslie R. Bridges
- Department of Cellular PathologySt George's University Hospitals NHS Foundation TrustLondonUK
| | - Margaret M. Esiri
- Nuffield Department of Clinical NeurosciencesOxford UniversityOxfordUK
| | - Joanne Lam‐Wong
- Department of Cellular PathologySt George's University Hospitals NHS Foundation TrustLondonUK
| | - James A. R. Nicoll
- Clinical Neurosciences, Clinical & Experimental SciencesUniversity of SouthamptonSouthamptonUK
| | - Delphine Boche
- Clinical Neurosciences, Clinical & Experimental SciencesUniversity of SouthamptonSouthamptonUK
| | - Atticus H. Hainsworth
- Neuroscience Research Centre, Molecular and Clinical Sciences Research InstituteSt. George’s, University of LondonLondonUK
| |
Collapse
|
26
|
Refolo V, Stefanova N. Neuroinflammation and Glial Phenotypic Changes in Alpha-Synucleinopathies. Front Cell Neurosci 2019; 13:263. [PMID: 31263402 PMCID: PMC6585624 DOI: 10.3389/fncel.2019.00263] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/28/2019] [Indexed: 01/10/2023] Open
Abstract
The role of neuroinflammation has been increasingly recognized in the field of neurodegenerative diseases. Many studies focusing on the glial cells involved in the inflammatory responses of the brain, namely microglia and astroglia, have over the years pointed out the dynamic and changing behavior of these cells, accompanied by different morphologies and activation forms. This is particularly evident in diseased conditions, where glia react to any shift from homeostasis, acquiring different phenotypes. Particularly for microglia, it has soon become clear that such phenotypes are multiple, as multiple are the functions related to them. Several approaches have over time revealed different facets of microglial phenotypic diversity, and advanced genetic analyses, in recent years, have added new insights into microglial heterogeneity, opening novel scenarios that researchers have just started to explore. Among neurodegenerative diseases, an important section is represented by alpha-synucleinopathies. Here alpha-synuclein accumulates abnormally in the brain and, depending on its pattern of distribution, leads to the development of different clinical conditions. Also for these proteinopathies, neuroinflammation and glial activation have been identified as constant and crucial factors during disease development. In the present review we will address the current literature about glial phenotypic changes with respect to alpha-synucleinopathies, as well as consider the pathophysiological and therapeutic implications of such a dynamic cellular behavior.
Collapse
Affiliation(s)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Gomes GF, Peixoto RDDF, Maciel BG, Santos KFD, Bayma LR, Feitoza Neto PA, Fernandes TN, de Abreu CC, Casseb SMM, de Lima CM, de Oliveira MA, Diniz DG, Vasconcelos PFDC, Sosthenes MCK, Diniz CWP. Differential Microglial Morphological Response, TNFα, and Viral Load in Sedentary-like and Active Murine Models After Systemic Non-neurotropic Dengue Virus Infection. J Histochem Cytochem 2019; 67:419-439. [PMID: 30924711 DOI: 10.1369/0022155419835218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Peripheral inflammatory stimuli increase proinflammatory cytokines in the bloodstream and central nervous system and activate microglial cells. Here we tested the hypothesis that contrasting environments mimicking sedentary and active lives would be associated with differential microglial morphological responses, inflammatory cytokines concentration, and virus load in the peripheral blood. For this, mice were maintained either in standard (standard environment) or enriched cages (enriched environment) and then subjected to a single (DENV1) serotype infection. Blood samples from infected animals showed higher viral loads and higher tumor necrosis factor-α (TNFα) mRNA concentrations than control subjects. Using an unbiased stereological sampling approach, we selected 544 microglia from lateral septum for microscopic 3D reconstruction. Morphological complexity contributed most to cluster formation. Infected groups exhibited significant increase in the microglia morphological complexity and number, despite the absence of dengue virus antigens in the brain. Two microglial phenotypes (type I with lower and type II with higher morphological complexity) were found in both infected and control groups. However, microglia from infected mice maintained in enriched environment showed only one morphological phenotype. Two-way ANOVA revealed that environmental changes and infection influenced type-I and II microglial morphologies and number. Environmental enrichment and infection interactions may contribute to microglial morphological change to a point that type-I and II morphological phenotypes could no longer be distinguished in infected mice from enriched environment. Significant linear correlation was found between morphological complexity and TNFα peripheral blood. Our findings demonstrated that sedentary-like and active murine models exhibited differential microglial responses and peripheral inflammation to systemic non-neurotropic infections with DENV1 virus.
Collapse
Affiliation(s)
- Giovanni Freitas Gomes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Railana Deise da Fonseca Peixoto
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Brenda Gonçalves Maciel
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Kedma Farias Dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Lohrane Rosa Bayma
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Pedro Alves Feitoza Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Taiany Nogueira Fernandes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Cintya Castro de Abreu
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | | | - Camila Mendes de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Marcus Augusto de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | | | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brasil
| |
Collapse
|
28
|
Hultman K, Scarlett JM, Baquero AF, Cornea A, Zhang Y, Salinas CBG, Brown J, Morton GJ, Whalen EJ, Grove KL, Koegler FH, Schwartz MW, Mercer AJ. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J Comp Neurol 2019; 527:2069-2085. [PMID: 30809795 DOI: 10.1002/cne.24668] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/25/2022]
Abstract
Central activation of fibroblast growth factor (FGF) receptors regulates peripheral glucose homeostasis and reduces food intake in preclinical models of obesity and diabetes. The current work was undertaken to advance our understanding of the receptor expression, as sites of ligand action by FGF19, FGF21, and FGF1 in the mammalian brain remains unresolved. Recent advances in automated RNAscope in situ hybridization and droplet digital PCR (ddPCR) technology allowed us to interrogate central FGFR/beta klotho (Klb) system at the cellular level in the mouse, with relevant comparisons to nonhuman primate and human brain. FGFR1-3 gene expression was broadly distributed throughout the CNS in Mus musculus, with FGFR1 exhibiting the greatest heterogeneity. FGFR4 expression localized only in the medial habenula and subcommissural organ of mice. Likewise, Klb mRNA was restricted to the suprachiasmatic nucleus (SCh) and select midbrain and hindbrain nuclei. ddPCR in the rodent hypothalamus confirmed that, although expression levels are indeed low for Klb, there is nonetheless a bonafide subpopulation of Klb+ cells in the hypothalamus. In NHP and human midbrain and hindbrain, Klb + cells are quite rare, as is expression of FGFR4. Collectively, these data provide the most robust central map of the FGFR/Klb system to date and highlight central regions that may be of critical importance to assess central ligand effects with pharmacological dosing, such as the putative interactions between the endocrine FGFs and FGFR1/Klb, or FGF19 with FGFR4.
Collapse
Affiliation(s)
| | - Jarrad M Scarlett
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington.,Department of Pediatric Gastroenterology & Hepatology, Seattle Children's Hospital, Seattle, Washington
| | - Arian F Baquero
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Anda Cornea
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Yu Zhang
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | | | - Jenny Brown
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Gregory J Morton
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Erin J Whalen
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Kevin L Grove
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Frank H Koegler
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Michael W Schwartz
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Aaron J Mercer
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| |
Collapse
|
29
|
Fiore NT, Austin PJ. Glial-cytokine-neuronal Adaptations in the Ventral Hippocampus of Rats with Affective Behavioral Changes Following Peripheral Nerve Injury. Neuroscience 2018; 390:119-140. [DOI: 10.1016/j.neuroscience.2018.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
|
30
|
SIRT1 activation with neuroheal is neuroprotective but SIRT2 inhibition with AK7 is detrimental for disconnected motoneurons. Cell Death Dis 2018; 9:531. [PMID: 29748539 PMCID: PMC5945655 DOI: 10.1038/s41419-018-0553-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/24/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023]
Abstract
Sirtuin 1 (SIRT1) activity is neuroprotective, and we have recently demonstrated its role in the retrograde degenerative process in motoneurons (MNs) in the spinal cord of rats after peripheral nerve root avulsion (RA) injury. SIRT2 has been suggested to exert effects opposite those of SIRT1; however, its roles in neurodegeneration and neuron response after nerve injury remain unclear. Here we compared the neuroprotective potentials of SIRT1 activation and SIRT2 inhibition in a mouse model of hypoglossal nerve axotomy. This injury induced a reduction of around half MN population within the hypoglossal nucleus by a non-apoptotic neurodegenerative process triggered by endoplasmic reticulum (ER) stress that resulted in activation of the unfolded protein response mediated by IRE1α and XBP1 by 21 days post injury. Both SIRT1 activation with NeuroHeal and SIRT2 inhibition with AK7 protected NSC-34 motor neuron-like cells against ER stress in vitro. In agreement with the in vitro results, NeuroHeal treatment or SIRT1 overexpression was neuroprotective of axotomized hypoglossal MNs in a transgenic mouse model. In contrast, AK7 treatment or SIRT2 genetic depletion in mice inhibited damaged MN survival. To resolve the in vitro/in vivo discrepancies, we used an organotypic spinal cord culture system that preserves glial cells. In this system, AK7 treatment of ER-stressed organotypic cultures was detrimental for MNs and increased microglial nuclear factor-κB and the consequent transcription of cytotoxic pro-inflammatory factors similarly. The results highlight the importance of glial cells in determining the neuroprotective impact of any treatment.
Collapse
|
31
|
Lanjakornsiripan D, Pior BJ, Kawaguchi D, Furutachi S, Tahara T, Katsuyama Y, Suzuki Y, Fukazawa Y, Gotoh Y. Layer-specific morphological and molecular differences in neocortical astrocytes and their dependence on neuronal layers. Nat Commun 2018; 9:1623. [PMID: 29691400 PMCID: PMC5915416 DOI: 10.1038/s41467-018-03940-3] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 03/23/2018] [Indexed: 12/22/2022] Open
Abstract
Non-pial neocortical astrocytes have historically been thought to comprise largely a nondiverse population of protoplasmic astrocytes. Here we show that astrocytes of the mouse somatosensory cortex manifest layer-specific morphological and molecular differences. Two- and three-dimensional observations revealed that astrocytes in the different layers possess distinct morphologies as reflected by differences in cell orientation, territorial volume, and arborization. The extent of ensheathment of synaptic clefts by astrocytes in layer II/III was greater than that by those in layer VI. Moreover, differences in gene expression were observed between upper-layer and deep-layer astrocytes. Importantly, layer-specific differences in astrocyte properties were abrogated in reeler and Dab1 conditional knockout mice, in which neuronal layers are disturbed, suggesting that neuronal layers are a prerequisite for the observed morphological and molecular differences of neocortical astrocytes. This study thus demonstrates the existence of layer-specific interactions between neurons and astrocytes, which may underlie their layer-specific functions.
Collapse
Affiliation(s)
- Darin Lanjakornsiripan
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Baek-Jun Pior
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Daichi Kawaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Shohei Furutachi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan. .,Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, W1T 4JG, UK.
| | - Tomoaki Tahara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Yutaka Suzuki
- Department of Computational Biology, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 277-8561, Japan
| | - Yugo Fukazawa
- Graduate School of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
32
|
Primary Traumatic Axonopathy in Mice Subjected to Impact Acceleration: A Reappraisal of Pathology and Mechanisms with High-Resolution Anatomical Methods. J Neurosci 2018; 38:4031-4047. [PMID: 29567804 DOI: 10.1523/jneurosci.2343-17.2018] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 02/21/2018] [Accepted: 03/15/2018] [Indexed: 11/21/2022] Open
Abstract
Traumatic axonal injury (TAI) is a common neuropathology in traumatic brain injury and is featured by primary injury to axons. Here, we generated TAI with impact acceleration of the head in male Thy1-eYFP-H transgenic mice in which specific populations of neurons and their axons are labeled with yellow fluorescent protein. This model results in axonal lesions in multiple axonal tracts along with blood-brain barrier disruption and neuroinflammation. The corticospinal tract, a prototypical long tract, is severely affected and is the focus of this study. Using optimized CLARITY at single-axon resolution, we visualized the entire corticospinal tract volume from the pons to the cervical spinal cord in 3D and counted the total number of axonal lesions and their progression over time. Our results divulged the presence of progressive traumatic axonopathy that was maximal at the pyramidal decussation. The perikarya of injured corticospinal neurons atrophied, but there was no evidence of neuronal cell death. We also used CLARITY at single-axon resolution to explore the role of the NMNAT2-SARM1 axonal self-destruction pathway in traumatic axonopathy. When we interfered with this pathway by genetically ablating SARM1 or by pharmacological strategies designed to increase levels of Nicotinamide (Nam), a feedback inhibitor of SARM1, we found a significant reduction in the number of axonal lesions early after injury. Our findings show that high-resolution neuroanatomical strategies reveal important features of TAI with biological implications, especially the progressive axonopathic nature of TAI and the role of the NMNAT2-SARM1 pathway in the early stages of axonopathy.SIGNIFICANCE STATEMENT In the first systematic application of novel high-resolution neuroanatomical tools in neuropathology, we combined CLARITY with 2-photon microscopy, optimized for detection of single axonal lesions, to reconstruct the injured mouse brainstem in a model of traumatic axonal injury (TAI) that is a common pathology associated with traumatic brain injury. The 3D reconstruction of the corticospinal tract at single-axon resolution allowed for a more advanced level of qualitative and quantitative understanding of TAI. Using this model, we showed that TAI is an axonopathy with a prominent role of the NMNAT2-SARM1 molecular pathway, that is also implicated in peripheral neuropathy. Our results indicate that high-resolution anatomical models of TAI afford a level of detail and sensitivity that is ideal for testing novel molecular and biomechanical hypotheses.
Collapse
|
33
|
Cătălin B, Stopper L, Bălşeanu TA, Scheller A. The in situ morphology of microglia is highly sensitive to the mode of tissue fixation. J Chem Neuroanat 2017; 86:59-66. [PMID: 28866082 DOI: 10.1016/j.jchemneu.2017.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/20/2017] [Accepted: 08/29/2017] [Indexed: 01/11/2023]
Abstract
Microglia are known as the most motile cells in the central nervous system (CNS). It was shown in vivo that they permanently scan their direct microenvironment and react to pathological conditions within minutes. Many studies of brain pathologies use fixed brain tissue to investigate cellular changes. Unfortunately, due to technical reasons, the time span between the induction of the fixation procedure (start of the perfusion) and the finally-fixed tissue lasts several minutes, giving time to microglia to start reacting to the ischemic conditions due to perfusion start. Here, we investigated the microglial changes generated by the fixation itself in TgH(CX3CR1-EGFP) mice with fluorescent labelled microglia using confocal laser scanning microscopy (CLSM) of fixed brain tissue as well as two-photon laser scanning microscopy (2P-LSM) during the perfusion of a living animal. We revealed the impact of fixation and buffer parameters on cell morphology. The largest morphological differences compared to physiological in vivo branch arborization were observed when the directly dissected brain was immersed in paraformaldehyde fixation solution overnight, without prior fixative perfusion of the animal. But even perfusion with a fixative, followed by post-fixation leads to small changes in microglial process length and number and could not be prevented when compared to physiological in vivo microglia morphology acquired using in vivo 2P-LSM. Interestingly, perfusion with different buffers either oxygenated artificial cerebrospinal fluid or phosphate buffered saline prior to perfusion-fixation showed minor microglia changes in arborization and/or number of processes. Fixation methods influence microglia morphology. Therefore, to define microglia activation states immunohistochemical stainings or genetic labelling of the cells have to be included in addition to morphological analysis.
Collapse
Affiliation(s)
- Bogdan Cătălin
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Building 48, University of Saarland, Homburg, Germany; Department of Functional Sciences, University of Medicine and Pharmacy of Craiova, Romania.
| | - Laura Stopper
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Building 48, University of Saarland, Homburg, Germany
| | - Tudor-Adrian Bălşeanu
- Department of Functional Sciences, University of Medicine and Pharmacy of Craiova, Romania; Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy of Craiova, Romania
| | - Anja Scheller
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Building 48, University of Saarland, Homburg, Germany.
| |
Collapse
|
34
|
Anderson WD, Greenhalgh AD, Takwale A, David S, Vadigepalli R. Novel Influences of IL-10 on CNS Inflammation Revealed by Integrated Analyses of Cytokine Networks and Microglial Morphology. Front Cell Neurosci 2017; 11:233. [PMID: 28855862 PMCID: PMC5557777 DOI: 10.3389/fncel.2017.00233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/25/2017] [Indexed: 01/16/2023] Open
Abstract
Coordinated interactions between cytokine signaling and morphological dynamics of microglial cells regulate neuroinflammation in CNS injury and disease. We found that pro-inflammatory cytokine gene expression in vivo showed a pronounced recovery following systemic LPS. We performed a novel multivariate analysis of microglial morphology and identified changes in specific morphological properties of microglia that matched the expression dynamics of pro-inflammatory cytokine TNFα. The adaptive recovery kinetics of TNFα expression and microglial soma size showed comparable profiles and dependence on anti-inflammatory cytokine IL-10 expression. The recovery of cytokine variations and microglial morphology responses to inflammation were negatively regulated by IL-10. Our novel morphological analysis of microglia is able to detect subtle changes and can be used widely. We implemented in silico simulations of cytokine network dynamics which showed—counter-intuitively, but in line with our experimental observations—that negative feedback from IL-10 was sufficient to impede the adaptive recovery of TNFα-mediated inflammation. Our integrative approach is a powerful tool to study changes in specific components of microglial morphology for insights into their functional states, in relation to cytokine network dynamics, during CNS injury and disease.
Collapse
Affiliation(s)
- Warren D Anderson
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson UniversityPhiladelphia, PA, United States
| | - Andrew D Greenhalgh
- Center for Research in Neuroscience, The Research Institute of the McGill University Health CenterMontreal, QC, Canada
| | - Aditya Takwale
- Center for Research in Neuroscience, The Research Institute of the McGill University Health CenterMontreal, QC, Canada
| | - Samuel David
- Center for Research in Neuroscience, The Research Institute of the McGill University Health CenterMontreal, QC, Canada
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson UniversityPhiladelphia, PA, United States
| |
Collapse
|
35
|
Fernández-Arjona MDM, Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. Microglia Morphological Categorization in a Rat Model of Neuroinflammation by Hierarchical Cluster and Principal Components Analysis. Front Cell Neurosci 2017; 11:235. [PMID: 28848398 PMCID: PMC5550745 DOI: 10.3389/fncel.2017.00235] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022] Open
Abstract
It is known that microglia morphology and function are closely related, but only few studies have objectively described different morphological subtypes. To address this issue, morphological parameters of microglial cells were analyzed in a rat model of aseptic neuroinflammation. After the injection of a single dose of the enzyme neuraminidase (NA) within the lateral ventricle (LV) an acute inflammatory process occurs. Sections from NA-injected animals and sham controls were immunolabeled with the microglial marker IBA1, which highlights ramifications and features of the cell shape. Using images obtained by section scanning, individual microglial cells were sampled from various regions (septofimbrial nucleus, hippocampus and hypothalamus) at different times post-injection (2, 4 and 12 h). Each cell yielded a set of 15 morphological parameters by means of image analysis software. Five initial parameters (including fractal measures) were statistically different in cells from NA-injected rats (most of them IL-1β positive, i.e., M1-state) compared to those from control animals (none of them IL-1β positive, i.e., surveillant state). However, additional multimodal parameters were revealed more suitable for hierarchical cluster analysis (HCA). This method pointed out the classification of microglia population in four clusters. Furthermore, a linear discriminant analysis (LDA) suggested three specific parameters to objectively classify any microglia by a decision tree. In addition, a principal components analysis (PCA) revealed two extra valuable variables that allowed to further classifying microglia in a total of eight sub-clusters or types. The spatio-temporal distribution of these different morphotypes in our rat inflammation model allowed to relate specific morphotypes with microglial activation status and brain location. An objective method for microglia classification based on morphological parameters is proposed. Main pointsMicroglia undergo a quantifiable morphological change upon neuraminidase induced inflammation. Hierarchical cluster and principal components analysis allow morphological classification of microglia. Brain location of microglia is a relevant factor.
Collapse
Affiliation(s)
- María Del Mar Fernández-Arjona
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - Pablo Granados-Durán
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - Pedro Fernández-Llebrez
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| | - María D López-Ávalos
- Departamento de Biología Celular, Facultad de Ciencias, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de MálagaMálaga, Spain
| |
Collapse
|
36
|
Ohgomori T, Yamasaki R, Takeuchi H, Kadomatsu K, Kira JI, Jinno S. Differential activation of neuronal and glial STAT3 in the spinal cord of the SOD1 G93A mouse model of amyotrophic lateral sclerosis. Eur J Neurosci 2017; 46:2001-2014. [PMID: 28715117 DOI: 10.1111/ejn.13650] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
Signal transducer and activator of transcription (STAT) proteins are activated by phosphorylation in the spinal cord of patients suffering from amyotrophic lateral sclerosis (ALS). The major scope of our study is a comprehensive histological characterization of the mechanisms underlying neuronal and glial STAT3 activation in the pathogenesis of ALS using SOD1G93A mice. We calculated the fold changes (FCs, ratios vs. appropriate controls) of the numerical densities of the following phosphorylated STAT3-positive (pSTAT3)+ cells - choline acetyltransferase (ChAT)+ α-motoneurons, ionized calcium-binding adapter molecule 1 (Iba1)+ microglia, and S100β+ astrocytes in SOD1G93A mice. The FCs of pSTAT3+ microglia and pSTAT3+ astrocytes were increased from 9 to 15 weeks of age and then plateaued until 21 weeks. In contrast, the FCs of pSTAT3+ α-motoneurons peaked at 9 weeks and then decreased until 21 weeks. The immunoreactivity for nonphosphorylated neurofilament protein (SMI-32), a marker of axonal impairment, was decreased in pSTAT3+ α-motoneurons compared with pSTAT3- α-motoneurons at 9 weeks of age. We then compared the following pharmacological models - the chronic administration of 3,3'-iminodipropionitrile (IDPN), which models axonal impairment, and the acute administration of lipopolysaccharide (LPS), which is a model of neuroinflammation. The FCs of pSTAT3+ α-motoneurons were increased in IDPN-treated mice, while those of pSTAT3+ microglia were increased in LPS-treated mice. The FCs of pSTAT3+ astrocytes were higher in SOD1G93A mice at 9 weeks compared with IDPN- and LPS-treated mice. Our results indicate that axonopathy and neuroinflammation may trigger the respective activation of neuronal and glial STAT3, which is observed during ALS pathogenesis.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| |
Collapse
|
37
|
Ohgomori T, Yamada J, Takeuchi H, Kadomatsu K, Jinno S. Comparative morphometric analysis of microglia in the spinal cord of SOD1(G93A) transgenic mouse model of amyotrophic lateral sclerosis. Eur J Neurosci 2016; 43:1340-51. [PMID: 26946061 DOI: 10.1111/ejn.13227] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 12/13/2022]
Abstract
It has long been recognized that reactive microglia undergo a series of phenotypic changes accompanying morphological transformation. However, the morphological classification of microglia has not yet been achieved. To address this issue, here we morphometrically analysed three-dimensionally reconstructed ionized calcium binding adaptor molecule 1-immunoreactive (Iba1(+) ) microglia in the ventral horn of the lumbar spinal cord of SOD1(G93A) transgenic mice, a model of amyotrophic lateral sclerosis. The hierarchical cluster analysis revealed that microglia were objectively divided into four groups: type S (named after surveillant microglia) and types R1, R2 and R3 (named after reactive microglia). For the purpose of comparative morphometry, we also analysed two pharmacological disease models using wild-type mice: 3,3'-iminodipropionitrile (IDPN)-induced axonopathy and lipopolysaccharide (LPS)-induced neuroinflammation. Type S microglia showed a typical ramified morphology of surveillant microglia, and were mostly observed in wild-type controls. Type R1 microglia were seen at the early stage of disease in SOD1(G93A) mice, and also frequently occurred in IDPN-treated mice. They exhibited small cell bodies with shorter and simple processes. Type R2 microglia were morphologically similar to type R1 microglia, but only transiently occurred in the middle stage of disease in SOD1(G93A) mice and in IDPN-treated mice. Type R3 microglia exhibited a bushy shape, and were observed in the end stage of disease in SOD1(G93A) mice and in LPS-treated mice. These findings indicate that microglia of SOD1(G93A) mice can be classified into four types, and also suggest that the phenotypic changes may be induced by the events related to axonopathy and neuroinflammation.
Collapse
Affiliation(s)
- Tomohiro Ohgomori
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hideyuki Takeuchi
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
38
|
Diniz DG, Silva GO, Naves TB, Fernandes TN, Araújo SC, Diniz JAP, de Farias LHS, Sosthenes MCK, Diniz CG, Anthony DC, da Costa Vasconcelos PF, Picanço Diniz CW. Hierarchical Cluster Analysis of Three-Dimensional Reconstructions of Unbiased Sampled Microglia Shows not Continuous Morphological Changes from Stage 1 to 2 after Multiple Dengue Infections in Callithrix penicillata. Front Neuroanat 2016; 10:23. [PMID: 27047345 PMCID: PMC4801861 DOI: 10.3389/fnana.2016.00023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/23/2016] [Indexed: 11/18/2022] Open
Abstract
It is known that microglial morphology and function are related, but few studies have explored the subtleties of microglial morphological changes in response to specific pathogens. In the present report we quantitated microglia morphological changes in a monkey model of dengue disease with virus CNS invasion. To mimic multiple infections that usually occur in endemic areas, where higher dengue infection incidence and abundant mosquito vectors carrying different serotypes coexist, subjects received once a week subcutaneous injections of DENV3 (genotype III)-infected culture supernatant followed 24 h later by an injection of anti-DENV2 antibody. Control animals received either weekly anti-DENV2 antibodies, or no injections. Brain sections were immunolabeled for DENV3 antigens and IBA-1. Random and systematic microglial samples were taken from the polymorphic layer of dentate gyrus for 3-D reconstructions, where we found intense immunostaining for TNFα and DENV3 virus antigens. We submitted all bi- or multimodal morphological parameters of microglia to hierarchical cluster analysis and found two major morphological phenotypes designated types I and II. Compared to type I (stage 1), type II microglia were more complex; displaying higher number of nodes, processes and trees and larger surface area and volumes (stage 2). Type II microglia were found only in infected monkeys, whereas type I microglia was found in both control and infected subjects. Hierarchical cluster analysis of morphological parameters of 3-D reconstructions of random and systematic selected samples in control and ADE dengue infected monkeys suggests that microglia morphological changes from stage 1 to stage 2 may not be continuous.
Collapse
Affiliation(s)
- Daniel G Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros BarretoBelém, Brasil; Experimental Neuropathology Laboratory, Department of Pharmacology, University of OxfordOxford, UK
| | - Geane O Silva
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto Belém, Brasil
| | - Thaís B Naves
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto Belém, Brasil
| | | | - Sanderson C Araújo
- Departamento de Microscopia Eletrônica, Instituto Evandro Chagas Belém, Brasil
| | - José A P Diniz
- Departamento de Microscopia Eletrônica, Instituto Evandro Chagas Belém, Brasil
| | - Luis H S de Farias
- Instituto de Ciências Biológicas, Universidade Federal do Pará Belém, Brasil
| | - Marcia C K Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto Belém, Brasil
| | - Cristovam G Diniz
- Laboratório de Biologia Molecular e Ambiental, Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus de Bragança, Bragança Pará, Brasil
| | - Daniel C Anthony
- Experimental Neuropathology Laboratory, Department of Pharmacology, University of Oxford Oxford, UK
| | | | - Cristovam W Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros BarretoBelém, Brasil; Experimental Neuropathology Laboratory, Department of Pharmacology, University of OxfordOxford, UK
| |
Collapse
|
39
|
Xu L, Schaefer ML, Linville RM, Aggarwal A, Mbuguiro W, Wester BA, Koliatsos VE. Neuroinflammation in primary blast neurotrauma: Time course and prevention by torso shielding. Exp Neurol 2016; 277:268-274. [DOI: 10.1016/j.expneurol.2016.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/02/2015] [Accepted: 01/13/2016] [Indexed: 01/12/2023]
|
40
|
Xu L, Nguyen JV, Lehar M, Menon A, Rha E, Arena J, Ryu J, Marsh-Armstrong N, Marmarou CR, Koliatsos VE. Repetitive mild traumatic brain injury with impact acceleration in the mouse: Multifocal axonopathy, neuroinflammation, and neurodegeneration in the visual system. Exp Neurol 2016; 275 Pt 3:436-449. [DOI: 10.1016/j.expneurol.2014.11.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/29/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
|
41
|
Vasconcelos BCB, Vieira JA, Silva GO, Fernandes TN, Rocha LC, Viana AP, Serique CDS, Filho CS, Bringel RAR, Teixeira FFDL, Ferreira MS, Casseb SMM, Carvalho VL, de Melo KFL, de Castro PHG, Araújo SC, Diniz JAP, Demachki S, Anaissi AKM, Sosthenes MCK, Vasconcelos PFDC, Anthony DC, Diniz CWP, Diniz DG. Antibody-enhanced dengue disease generates a marked CNS inflammatory response in the black-tufted marmoset Callithrix penicillata. Neuropathology 2015; 36:3-16. [PMID: 26303046 DOI: 10.1111/neup.12229] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
Severe dengue disease is often associated with long-term neurological impairments, but it is unclear what mechanisms are associated with neurological sequelae. Previously, we demonstrated antibody-enhanced dengue disease (ADE) dengue in an immunocompetent mouse model with a dengue virus 2 (DENV2) antibody injection followed by DENV3 virus infection. Here we migrated this ADE model to Callithrix penicillata. To mimic human multiple infections of endemic zones where abundant vectors and multiple serotypes co-exist, three animals received weekly subcutaneous injections of DENV3 (genotype III)-infected supernatant of C6/36 cell cultures, followed 24 h later by anti-DENV2 antibody for 12 weeks. There were six control animals, two of which received weekly anti-DENV2 antibodies, and four further animals received no injections. After multiple infections, brain, liver, and spleen samples were collected and tissue was immunolabeled for DENV3 antigens, ionized calcium binding adapter molecule 1, Ki-67, TNFα. There were marked morphological changes in the microglial population of ADE monkeys characterized by more highly ramified microglial processes, higher numbers of trees and larger surface areas. These changes were associated with intense TNFα-positive immunolabeling. It is unclear why ADE should generate such microglial activation given that IgG does not cross the blood-brain barrier, but this study reveals that in ADE dengue therapy targeting the CNS host response is likely to be important.
Collapse
Affiliation(s)
| | - Juliana Almeida Vieira
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - Geane Oliveira Silva
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | | | - Luciano Chaves Rocha
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - André Pereira Viana
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - Cássio Diego Sá Serique
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - Carlos Santos Filho
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - Raissa Aires Ribeiro Bringel
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - Francisco Fernando Dacier Lobato Teixeira
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | | | | | | | | | | | | | | | - Samia Demachki
- Universidade Federal do Pará, UFPA, Instituto de Ciências da Saúde, Laboratório de Anatomia Patológica, Hospital Universitário João de Barros Barreto, Belém, Pará, Brasil
| | - Ana Karyssa Mendes Anaissi
- Universidade Federal do Pará, UFPA, Instituto de Ciências da Saúde, Laboratório de Anatomia Patológica, Hospital Universitário João de Barros Barreto, Belém, Pará, Brasil
| | - Marcia Consentino Kronka Sosthenes
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | | | - Daniel Clive Anthony
- Department of Pharmacology, Laboratory of Experimental Neuropathology, University of Oxford, Mansfield Road, Oxford, United Kingdom
| | - Cristovam Wanderley Picanço Diniz
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| | - Daniel Guerreiro Diniz
- Universidade Federal do Pará, UFPA, Instituto de Ciências Biológicas, Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto
| |
Collapse
|
42
|
Gerenu G, Liu K, Chojnacki JE, Saathoff JM, Martínez-Martín P, Perry G, Zhu X, Lee HG, Zhang S. Curcumin/melatonin hybrid 5-(4-hydroxy-phenyl)-3-oxo-pentanoic acid [2-(5-methoxy-1H-indol-3-yl)-ethyl]-amide ameliorates AD-like pathology in the APP/PS1 mouse model. ACS Chem Neurosci 2015; 6:1393-1399. [PMID: 25893520 DOI: 10.1021/acschemneuro.5b00082] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In our efforts to develop hybrid compounds of curcumin and melatonin as potential disease-modifying agents for Alzheimer's disease (AD), a potent lead hybrid compound, Z-CM-I-1, has been recently identified and biologically characterized in vitro. In this work, we report the in vivo effects of Z-CM-I-1 on AD pathologies in an APP/PS1 transgenic AD model. Our studies demonstrated that Z-CM-I-1 significantly decreased the accumulation of Aβ in the hippocampus and cortex regions of the brain and reduced inflammatory responses and oxidative stress after treatment for 12 weeks at 50 mg/kg per dose via oral administration. Furthermore, Z-CM-I-1 significantly improved synaptic dysfunction evidenced by the increased expression of synaptic marker proteins, PSD95 and synaptophysin, indicating its protective effects on synaptic degeneration. Lastly, we demonstrated that Z-CM-I-1 significantly increased the expression level of complexes I, II, and IV of the mitochondria electron transport chain in the brain tissue of APP/PS1 mice. Collectively, these results clearly suggest that Z-CM-I-1 is orally available and exhibits multifunctional properties in vivo on AD pathologies, thus strongly encouraging further development of this lead compound as a potential disease-modifying agent for AD patients.
Collapse
Affiliation(s)
| | - Kai Liu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Jeremy E. Chojnacki
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - John M. Saathoff
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Pablo Martínez-Martín
- Alzheimer Disease Research Unit, Alzheimer Center Reina Sofía Foundation, CIEN Foundation, Carlos III Institute of Health, 28071 Madrid, Spain
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | | | | | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
43
|
de Sousa AA, Dos Reis RR, de Lima CM, de Oliveira MA, Fernandes TN, Gomes GF, Diniz DG, Magalhães NM, Diniz CG, Sosthenes MCK, Bento-Torres J, Diniz JAP, Vasconcelos PFDC, Diniz CWP. Three-dimensional morphometric analysis of microglial changes in a mouse model of virus encephalitis: age and environmental influences. Eur J Neurosci 2015; 42:2036-50. [PMID: 25980955 DOI: 10.1111/ejn.12951] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 04/16/2015] [Accepted: 05/13/2015] [Indexed: 02/05/2023]
Abstract
Many RNA virus CNS infections cause neurological disease. Because Piry virus has a limited human pathogenicity and exercise reduces activation of microglia in aged mice, possible influences of environment and aging on microglial morphology and behavior in mice sublethal encephalitis were investigated. Female albino Swiss mice were raised either in standard (S) or in enriched (EE) cages from age 2 to 6 months (young - Y), or from 2 to 16 months (aged - A). After behavioral tests, mice nostrils were instilled with Piry-virus-infected or with normal brain homogenates. Brain sections were immunolabeled for virus antigens or microglia at 8 days post-infection (dpi), when behavioral changes became apparent, and at 20 and 40 dpi, after additional behavioral testing. Young infected mice from standard (SYPy) and enriched (EYPy) groups showed similar transient impairment in burrowing activity and olfactory discrimination, whereas aged infected mice from both environments (EAPy, SAPy) showed permanent reduction in both tasks. The beneficial effects of an enriched environment were smaller in aged than in young mice. Six-hundred and forty microglial cells, 80 from each group were reconstructed. An unbiased, stereological sampling approach and multivariate statistical analysis were used to search for microglial morphological families. This procedure allowed distinguishing between microglial morphology of infected and control subjects. More severe virus-associated microglial changes were observed in young than in aged mice, and EYPy seem to recover microglial homeostatic morphology earlier than SYPy . Because Piry-virus encephalitis outcomes were more severe in aged mice, it is suggested that the reduced inflammatory response in those individuals may aggravate encephalitis outcomes.
Collapse
Affiliation(s)
- Aline A de Sousa
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Renata R Dos Reis
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Camila M de Lima
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Marcus A de Oliveira
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | | | - Giovanni F Gomes
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Daniel G Diniz
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Nara M Magalhães
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - Cristovam G Diniz
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Bragança, Pará, Brazil
| | - Marcia C K Sosthenes
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - João Bento-Torres
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil
| | - José Antonio P Diniz
- Instituto Evandro Chagas (IEC), Departamento de Arbovirologia e Febres Hemorrágicas, Ananindeua, Pará, Brazil
| | - Pedro F da C Vasconcelos
- Instituto Evandro Chagas (IEC), Departamento de Arbovirologia e Febres Hemorrágicas, Ananindeua, Pará, Brazil
| | - Cristovam Wanderley P Diniz
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Laboratório de Investigações em Neurodegeneração e Infecção no Hospital Universitário João de Barros Barreto, Belém, Pará, Brazil.,Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| |
Collapse
|
44
|
Role of Microglial Activation in the Pathophysiology of Bacterial Meningitis. Mol Neurobiol 2015; 53:1770-1781. [DOI: 10.1007/s12035-015-9107-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/20/2015] [Indexed: 12/18/2022]
|
45
|
Dennis CV, Sheahan PJ, Graeber MB, Sheedy DL, Kril JJ, Sutherland GT. Microglial proliferation in the brain of chronic alcoholics with hepatic encephalopathy. Metab Brain Dis 2014; 29:1027-39. [PMID: 24346482 PMCID: PMC4063896 DOI: 10.1007/s11011-013-9469-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/04/2013] [Indexed: 12/11/2022]
Abstract
Hepatic encephalopathy (HE) is a common complication of chronic alcoholism and patients show neurological symptoms ranging from mild cognitive dysfunction to coma and death. The HE brain is characterized by glial changes, including microglial activation, but the exact pathogenesis of HE is poorly understood. During a study investigating cell proliferation in the subventricular zone of chronic alcoholics, a single case with widespread proliferation throughout their adjacent grey and white matter was noted. This case also had concomitant HE raising the possibility that glial proliferation might be a pathological feature of the disease. In order to explore this possibility fixed postmortem human brain tissue from chronic alcoholics with cirrhosis and HE (n = 9), alcoholics without HE (n = 4) and controls (n = 4) were examined using immunohistochemistry and cytokine assays. In total, 4/9 HE cases had PCNA- and a second proliferative marker, Ki-67-positive cells throughout their brain and these cells co-stained with the microglial marker, Iba1. These cases were termed 'proliferative HE' (pHE). The microglia in pHEs displayed an activated morphology with hypertrophied cell bodies and short, thickened processes. In contrast, the microglia in white matter regions of the non-proliferative HE cases were less activated and appeared dystrophic. pHEs were also characterized by higher interleukin-6 levels and a slightly higher neuronal density . These findings suggest that microglial proliferation may form part of an early neuroprotective response in HE that ultimately fails to halt the course of the disease because underlying etiological factors such as high cerebral ammonia and systemic inflammation remain.
Collapse
Affiliation(s)
- Claude V Dennis
- Discipline of Pathology, Sydney Medical School, Camperdown, NSW, 2050, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Tello Velasquez J, Watts ME, Todorovic M, Nazareth L, Pastrana E, Diaz-Nido J, Lim F, Ekberg JAK, Quinn RJ, John JAS. Low-dose curcumin stimulates proliferation, migration and phagocytic activity of olfactory ensheathing cells. PLoS One 2014; 9:e111787. [PMID: 25360677 PMCID: PMC4216124 DOI: 10.1371/journal.pone.0111787] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/30/2014] [Indexed: 12/22/2022] Open
Abstract
One of the promising strategies for neural repair therapies is the transplantation of olfactory ensheathing cells (OECs) which are the glial cells of the olfactory system. We evaluated the effects of curcumin on the behaviour of mouse OECs to determine if it could be of use to further enhance the therapeutic potential of OECs. Curcumin, a natural polyphenol compound found in the spice turmeric, is known for its anti-cancer properties at doses over 10 µM, and often at 50 µM, and it exerts its effects on cancer cells in part by activation of MAP kinases. In contrast, we found that low-dose curcumin (0.5 µM) applied to OECs strikingly modulated the dynamic morphology, increased the rate of migration by up to 4-fold, and promoted significant proliferation of the OECs. Most dramatically, low-dose curcumin stimulated a 10-fold increase in the phagocytic activity of OECs. All of these potently stimulated behavioural characteristics of OECs are favourable for neural repair therapies. Importantly, low-dose curcumin gave a transient activation of p38 kinases, which is in contrast to the high dose curcumin effects on cancer cells in which these MAP kinases tend to undergo prolonged activation. Low-dose curcumin mediated effects on OECs demonstrate cell-type specific stimulation of p38 and ERK kinases. These results constitute the first evidence that low-dose curcumin can modulate the behaviour of olfactory glia into a phenotype potentially more favourable for neural repair and thereby improve the therapeutic use of OECs for neural repair therapies.
Collapse
Affiliation(s)
| | - Michelle E. Watts
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - Michael Todorovic
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - Lynnmaria Nazareth
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Erika Pastrana
- Nature Communications, New York, New York, United States of America
| | | | - Filip Lim
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Jenny A. K. Ekberg
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Ronald J. Quinn
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - James A. St John
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Australia
- * E-mail:
| |
Collapse
|
47
|
Kongsui R, Beynon SB, Johnson SJ, Walker FR. Quantitative assessment of microglial morphology and density reveals remarkable consistency in the distribution and morphology of cells within the healthy prefrontal cortex of the rat. J Neuroinflammation 2014; 11:182. [PMID: 25343964 PMCID: PMC4213482 DOI: 10.1186/s12974-014-0182-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 10/10/2014] [Indexed: 11/24/2022] Open
Abstract
Background Microglial morphology within the healthy brain has been the subject of a number of observational studies. These have suggested that microglia may consist of separate classes, which possess substantially different morphological features. Critically, there have been no systematic quantitative studies of microglial morphology within the healthy brain. Methods We examined microglial cells within the adult rat prefrontal cortex. At high magnification, digital reconstructions of cells labelled with the microglial-specific marker ionized calcium-binding adapter molecule-1 (Iba-1) were made in each of the cortical layers. These reconstructions were subsequently analyzed to determine the convex hull area of the cells, their somal perimeter, the length of processes, the number of processes, the extent of process branching and the volume of processes. We additionally examined whether cells’ morphological features were associated with cell size or numerical density. Results Our analysis indicated that while there was substantial variability in the size of cells within the prefrontal cortex, cellular morphology was extremely consistent within each of the cortical layers. Conclusions Our results provide quantitative confirmation that microglia are largely homogenous in the uninjured rodent prefrontal cortex.
Collapse
|
48
|
Walker FR, Beynon SB, Jones KA, Zhao Z, Kongsui R, Cairns M, Nilsson M. Dynamic structural remodelling of microglia in health and disease: a review of the models, the signals and the mechanisms. Brain Behav Immun 2014; 37:1-14. [PMID: 24412599 DOI: 10.1016/j.bbi.2013.12.010] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 12/11/2013] [Accepted: 12/12/2013] [Indexed: 01/14/2023] Open
Abstract
Microglia are unique cells within the central nervous system because of their biophysical independence. As a result of this unusual property the cells must undergo significant structural remodelling in order to engage and connect with other elements within the central nervous system. Efficient remodelling is required for all activities that microglia are involved in ranging from monitoring synaptic information flow through to phagocytosis of tissue debris. Despite the fact that morphological remodelling is a pre-requisite to all microglial activities, relatively little research has been undertaken on the topic. This review examines what is known about how microglia transform themselves during development, under physiological conditions in response to changes in neuronal activity, and under pathological circumstances. Specific attention is given to exploring a variety of models that have been proposed to account for microglial transformation as well as the signals that are known to trigger these transformations.
Collapse
Affiliation(s)
- F Rohan Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Sarah B Beynon
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kimberley A Jones
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Zidan Zhao
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ratchaniporn Kongsui
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Murray Cairns
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Michael Nilsson
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Australia; Centre for Translational Neuroscience and Mental Health Research, University of Newcastle, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| |
Collapse
|