51
|
Welser JV, Halder SK, Kant R, Boroujerdi A, Milner R. Endothelial α6β4 integrin protects during experimental autoimmune encephalomyelitis-induced neuroinflammation by maintaining vascular integrity and tight junction protein expression. J Neuroinflammation 2017; 14:217. [PMID: 29121970 PMCID: PMC5679365 DOI: 10.1186/s12974-017-0987-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/24/2017] [Indexed: 12/23/2022] Open
Abstract
Background Extracellular matrix (ECM) proteins play critical functions regulating vascular formation and function. Laminin is a major component of the vascular basal lamina, and transgenic mice deficient in astrocyte or pericyte laminin show defective blood-brain barrier (BBB) integrity, indicating an important instructive role for laminin in cerebral blood vessels. As previous work shows that in the normal brain, vascular expression of the laminin receptor α6β4 integrin is predominantly restricted to arterioles, but induced on all vessels during neuroinflammation, it is important to define the role of this integrin in the maintenance of BBB integrity. Methods α6β4 integrin expression was analyzed using dual immunofluorescence (dual-IF) of brain sections taken from the mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). To investigate the role of endothelial α6β4 integrin, transgenic mice lacking β4 integrin in endothelial cells (β4-EC-KO) and wild-type (WT) littermates were subject to EAE, and clinical score and various neuropathological parameters were examined by immunofluorescence. In addition, β4 integrin null brain endothelial cells (BECs) were examined in culture for expression of tight junction proteins using immunocytochemistry and flow cytometry. Results Cerebrovascular expression of β4 integrin was markedly upregulated during EAE progression, such that by the acute stage of EAE (day 21), the vast majority of blood vessels expressed β4 integrin. In the EAE model, while the β4-EC-KO mice showed the same time of disease onset as the WT littermates, they developed significantly worse clinical disease over time, resulting in increased clinical score at the peak of disease and maintained elevated thereafter. Consistent with this, the β4-EC-KO mice showed enhanced levels of leukocyte infiltration and BBB breakdown and also displayed increased loss of the endothelial tight junction proteins claudin-5 and ZO-1. Under pro-inflammatory conditions, primary cultures of β4KO BECs also showed increased loss of claudin-5 and ZO-1 expression. Conclusions Taken together, our data suggest that α6β4 integrin upregulation is an inducible protective mechanism that stabilizes the BBB during neuroinflammatory conditions.
Collapse
Affiliation(s)
- Jennifer V Welser
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM-132, La Jolla, CA, 92037, USA
| | - Sebok K Halder
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM-132, La Jolla, CA, 92037, USA
| | - Ravi Kant
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM-132, La Jolla, CA, 92037, USA
| | - Amin Boroujerdi
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM-132, La Jolla, CA, 92037, USA
| | - Richard Milner
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MEM-132, La Jolla, CA, 92037, USA.
| |
Collapse
|
52
|
Biswas S, Bachay G, Chu J, Hunter DD, Brunken WJ. Laminin-Dependent Interaction between Astrocytes and Microglia: A Role in Retinal Angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2112-2127. [PMID: 28697326 DOI: 10.1016/j.ajpath.2017.05.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/25/2017] [Accepted: 05/31/2017] [Indexed: 11/18/2022]
Abstract
Retinal vascular diseases are among the leading causes of acquired blindness. In recent years, retinal microglia have been shown to influence vascular branching density and endothelial cell proliferation. However, how microglial recruitment and activation are regulated during development remains unclear. We hypothesized that microglial recruitment, activation, and down-stream signaling are modulated by components of the mural basement membrane. We used a reverse genetic approach to disrupt laminin expression in the vascular basement membrane and demonstrate that microglia respond to the mural basement membrane in an isoform-specific manner. Microglial density is significantly increased in the laminin γ3-null (Lamc3-/-) retinal superficial vascular plexus and consequently the vascular branching density is increased. Microglia also respond to astrocyte-derived matrices and become hyperactivated in the Lamc3-/- retina or when tested in vitro with cell-derived matrix. Pharmacological activation of microglia in the wild-type retina produced an Lamc3-/--like vascular phenotype, whereas pharmacological blocking of microglial activation in the Lamc3-/- retina rescued the wild-type vascular phenotype. On the molecular level, microglial transforming growth factor-β1 expression is down-regulated in the Lamc3-/- retina, and SMAD signaling decreased in endothelial cells with a consequent increase in endothelial proliferation. The reverse effects were seen in the Lamb2-/- retina. Together, our results demonstrate a novel mechanism by which laminins modulate vascular branching and endothelial cell proliferation during retinal angiogenesis.
Collapse
Affiliation(s)
- Saptarshi Biswas
- Department of Ophthalmology, State University of New York Upstate Medical University, Syracuse, New York; Department of Ophthalmology, State University of New York Downstate Medical Center, Brooklyn, New York; Center for Vision Research, Syracuse, New York
| | - Galina Bachay
- Department of Ophthalmology, State University of New York Upstate Medical University, Syracuse, New York; Department of Ophthalmology, State University of New York Downstate Medical Center, Brooklyn, New York; Center for Vision Research, Syracuse, New York
| | - Julianne Chu
- Department of Ophthalmology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Dale D Hunter
- Department of Ophthalmology, State University of New York Upstate Medical University, Syracuse, New York; Department of Ophthalmology, State University of New York Downstate Medical Center, Brooklyn, New York; Center for Vision Research, Syracuse, New York
| | - William J Brunken
- Department of Ophthalmology, State University of New York Upstate Medical University, Syracuse, New York; Department of Ophthalmology, State University of New York Downstate Medical Center, Brooklyn, New York; Center for Vision Research, Syracuse, New York.
| |
Collapse
|
53
|
Meller J, Chen Z, Dudiki T, Cull RM, Murtazina R, Bal SK, Pluskota E, Stefl S, Plow EF, Trapp BD, Byzova TV. Integrin-Kindlin3 requirements for microglial motility in vivo are distinct from those for macrophages. JCI Insight 2017; 2:93002. [PMID: 28570266 PMCID: PMC5453700 DOI: 10.1172/jci.insight.93002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/25/2017] [Indexed: 11/17/2022] Open
Abstract
Microglia play a critical role in the development and homeostasis of the CNS. While mobilization of microglia is critical for a number of pathologies, understanding of the mechanisms of their migration in vivo is limited and often based on similarities to macrophages. Kindlin3 deficiency as well as Kindlin3 mutations of integrin-binding sites abolish both integrin inside-out and outside-in signaling in microglia, thereby resulting in severe deficiencies in cell adhesion, polarization, and migration in vitro, which are similar to the defects observed in macrophages. In contrast, while Kindlin3 mutations impaired macrophage mobilization in vivo, they had no effect either on the population of microglia in the CNS during development or on mobilization of microglia and subsequent microgliosis in a model of multiple sclerosis. At the same time, acute microglial response to laser-induced injury was impaired by the lack of Kindlin3-integrin interactions. Based on 2-photon imaging of microglia in the brain, Kindlin3 is required for elongation of microglial processes toward the injury site and formation of phagosomes in response to brain injury. Thus, while Kindlin3 deficiency in human subjects is not expected to diminish the presence of microglia within CNS, it might delay the recovery process after injury, thereby exacerbating its complications.
Collapse
Affiliation(s)
| | - Zhihong Chen
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | - Bruce D Trapp
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
54
|
Smolders SMT, Swinnen N, Kessels S, Arnauts K, Smolders S, Le Bras B, Rigo JM, Legendre P, Brône B. Age-specific function of α5β1 integrin in microglial migration during early colonization of the developing mouse cortex. Glia 2017; 65:1072-1088. [PMID: 28417486 DOI: 10.1002/glia.23145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
Microglia, the immune cells of the central nervous system, take part in brain development and homeostasis. They derive from primitive myeloid progenitors that originate in the yolk sac and colonize the brain mainly through intensive migration. During development, microglial migration speed declines which suggests that their interaction with the microenvironment changes. However, the matrix-cell interactions allowing dispersion within the parenchyma are unknown. Therefore, we aimed to better characterize the migration behavior and to assess the role of matrix-integrin interactions during microglial migration in the embryonic brain ex vivo. We focused on microglia-fibronectin interactions mediated through the fibronectin receptor α5β1 integrin because in vitro work indirectly suggested a role for this ligand-receptor pair. Using 2-photon time-lapse microscopy on acute ex vivo embryonic brain slices, we found that migration occurs in a saltatory pattern and is developmentally regulated. Most importantly, there is an age-specific function of the α5β1 integrin during microglial cortex colonization. At embryonic day (E) 13.5, α5β1 facilitates migration while from E15.5, it inhibits migration. These results indicate a developmentally regulated function of α5β1 integrin in microglial migration during colonization of the embryonic brain.
Collapse
Affiliation(s)
- Sophie Marie-Thérèse Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium.,INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | | | | | - Silke Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium.,Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Barbara Le Bras
- INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | - Pascal Legendre
- INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | |
Collapse
|
55
|
Rodríguez AM, Delpino MV, Miraglia MC, Costa Franco MM, Barrionuevo P, Dennis VA, Oliveira SC, Giambartolomei GH. Brucella abortus-activated microglia induce neuronal death through primary phagocytosis. Glia 2017; 65:1137-1151. [PMID: 28398652 DOI: 10.1002/glia.23149] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/13/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Affiliation(s)
- Ana M. Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| | - M. Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| | - M. Cruz Miraglia
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| | - Miriam M. Costa Franco
- Department of Biochemistry and Immunology; Institute of Biological Sciences, Federal University of Minas Gerais; Belo Horizonte-Minas Gerais Brazil
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina); Buenos Aires Argentina
| | - Vida A. Dennis
- Center for Nano Biotechnology Research and Department of Biological Sciences; Alabama State University; Montgomery AL
| | - Sergio C. Oliveira
- Department of Biochemistry and Immunology; Institute of Biological Sciences, Federal University of Minas Gerais; Belo Horizonte-Minas Gerais Brazil
| | - Guillermo H. Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
56
|
Noor S, Sanchez JJ, Vanderwall AG, Sun MS, Maxwell JR, Davies S, Jantzie LL, Petersen TR, Savage DD, Milligan ED. Prenatal alcohol exposure potentiates chronic neuropathic pain, spinal glial and immune cell activation and alters sciatic nerve and DRG cytokine levels. Brain Behav Immun 2017; 61:80-95. [PMID: 28011263 PMCID: PMC5316367 DOI: 10.1016/j.bbi.2016.12.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/18/2016] [Accepted: 12/18/2016] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence indicates that prenatal alcohol exposure (PAE) may predispose individuals to secondary medical disabilities later in life. Animal models of PAE reveal neuroimmune sequelae such as elevated brain astrocyte and microglial activation with corresponding region-specific changes in immune signaling molecules such as cytokines and chemokines. The aim of this study was to evaluate the effects of moderate PAE on the development and maintenance of allodynia induced by chronic constriction injury (CCI) of the sciatic nerve in adult male rat offspring. Because CCI allodynia requires the actions of glial cytokines, we analyzed lumbar spinal cord glial and immune cell surface markers indicative of their activation levels, as well as sciatic nerve and dorsal root ganglia (DRG) cytokines in PAE offspring in adulthood. While PAE did not alter basal sensory thresholds before or after sham manipulations, PAE significantly potentiated adult onset and maintenance of allodynia. Microscopic analysis revealed exaggerated astrocyte and microglial activation, while flow cytometry data demonstrated increased proportions of immune cells with cell surface major histocompatibility complex II (MHCII) and β-integrin adhesion molecules, which are indicative of PAE-induced immune cell activation. Sciatic nerves from CCI rats revealed that PAE potentiated the proinflammatory cytokines interleukin (IL)-1β, IL-6 and tumor necrosis factor-alpha (TNFα) protein levels with a simultaneous robust suppression of the anti-inflammatory cytokine, IL-10. A profound reduction in IL-10 expression in the DRG of PAE neuropathic rats was also observed. Taken together, our results provide novel insights into the vulnerability that PAE produces for adult-onset central nervous system (CNS) pathological conditions from peripheral nerve injury.
Collapse
Affiliation(s)
- Shahani Noor
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Joshua J Sanchez
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Arden G Vanderwall
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA; Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Melody S Sun
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Jessie R Maxwell
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Suzy Davies
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Lauren L Jantzie
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA; Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Timothy R Petersen
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Daniel D Savage
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA; Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA
| | - Erin D Milligan
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA.
| |
Collapse
|
57
|
Eles JR, Vazquez AL, Snyder NR, Lagenaur C, Murphy MC, Kozai TDY, Cui XT. Neuroadhesive L1 coating attenuates acute microglial attachment to neural electrodes as revealed by live two-photon microscopy. Biomaterials 2016; 113:279-292. [PMID: 27837661 DOI: 10.1016/j.biomaterials.2016.10.054] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/26/2016] [Accepted: 10/30/2016] [Indexed: 12/15/2022]
Abstract
Implantable neural electrode technologies for chronic neural recordings can restore functional control to paralysis and limb loss victims through brain-machine interfaces. These probes, however, have high failure rates partly due to the biological responses to the probe which generate an inflammatory scar and subsequent neuronal cell death. L1 is a neuronal specific cell adhesion molecule and has been shown to minimize glial scar formation and promote electrode-neuron integration when covalently attached to the surface of neural probes. In this work, the acute microglial response to L1-coated neural probes was evaluated in vivo by implanting coated devices into the cortex of mice with fluorescently labeled microglia, and tracking microglial dynamics with multi-photon microscopy for the ensuing 6 h in order to understand L1's cellular mechanisms of action. Microglia became activated immediately after implantation, extending processes towards both L1-coated and uncoated control probes at similar velocities. After the processes made contact with the probes, microglial processes expanded to cover 47.7% of the control probes' surfaces. For L1-coated probes, however, there was a statistically significant 83% reduction in microglial surface coverage. This effect was sustained through the experiment. At 6 h post-implant, the radius of microglia activation was reduced for the L1 probes by 20%, shifting from 130.0 to 103.5 μm with the coating. Microglia as far as 270 μm from the implant site displayed significantly lower morphological characteristics of activation for the L1 group. These results suggest that the L1 surface treatment works in an acute setting by microglial mediated mechanisms.
Collapse
Affiliation(s)
- James R Eles
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States
| | - Alberto L Vazquez
- Bioengineering, University of Pittsburgh, United States; Radiology, University of Pittsburgh, United States; Neurobiology, University of Pittsburgh, United States
| | - Noah R Snyder
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States
| | - Carl Lagenaur
- Neurobiology, University of Pittsburgh, United States
| | | | - Takashi D Y Kozai
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, United States; NeuroTech Center of the University of Pittsburgh Brain Institute, United States.
| | - X Tracy Cui
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, United States.
| |
Collapse
|
58
|
The association between laminin and microglial morphology in vitro. Sci Rep 2016; 6:28580. [PMID: 27334934 PMCID: PMC4917827 DOI: 10.1038/srep28580] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 06/07/2016] [Indexed: 12/22/2022] Open
Abstract
Microglia are immune cells in the central nervous system (CNS) that contribute to primary innate immune responses. The morphology of microglia is closely associated with their functional activities. The majority of microglial studies have focused on the ramified or amoeboid morphology; however, bipolar/rod-shaped microglia have recently received much attention. Bipolar/rod-shaped microglia form trains with end-to-end alignment in injured brains and retinae, which is proposed as an important mechanism in CNS repair. We previously established a cell culture model system to enrich bipolar/rod-shaped microglia simply by growing primary microglia on scratched poly-D-lysine (PDL)/laminin-coated surfaces. Here, we investigated the role of laminin in morphological changes of microglia. Bipolar/rod-shaped microglia trains were transiently formed on scratched surfaces without PDL/laminin coating, but the microglia alignment disappeared after 3 days in culture. Amoeboid microglia digested the surrounding laminin, and the gene and protein expression of laminin-cleaving genes Adam9 and Ctss was up-regulated. Interestingly, lipopolysaccharide (LPS)-induced transformation from bipolar/rod-shaped into amoeboid microglia increased the expression of Adam9 and Ctss, and the expression of these genes in LPS-treated amoeboid-enriched cultures remained unchanged. These results indicate a strong association between laminin and morphological transformation of microglia, shedding new light on the role of bipolar/rod-shaped microglia in CNS repair.
Collapse
|
59
|
Udeochu JC, Shea JM, Villeda SA. Microglia communication: Parallels between aging and Alzheimer's disease. ACTA ACUST UNITED AC 2016; 7:114-125. [PMID: 27840659 PMCID: PMC5084774 DOI: 10.1111/cen3.12307] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 12/25/2022]
Abstract
Aging alters the functional integrity of the adult brain, driving cognitive impairments and susceptibility to neurodegenerative disorders in healthy individuals. In fact, aging remains the most dominant risk factor for Alzheimer's disease (AD). Recent findings have expanded our understanding of microglia function in the normal aging and AD brain, provoking an appreciation for microglia involvement in remodeling neuronal connections and maintaining brain integrity. This homeostatic function of microglia is achieved in part through the ability of microglia to interact extensively with and rapidly respond to changes in the brain microenvironment to enable adequate phenotypic transformations. Here, we discuss pro‐inflammatory drivers of microglia transformation in aging and AD by focusing on the immune‐modulatory functions of secreted factors, such as cytokines, complement factors and extracellular vesicles. We highlight the involvement of these secreted factors in aging and AD‐associated cellular changes in microglia immune activation, surveillance function, and phagocytosis. Finally, we discuss how pro‐inflammatory phenotypic changes associated with altered immune communication could both facilitate and exacerbate impairments in synaptic plasticity and cognitive function observed in the aged and AD brain.
Collapse
Affiliation(s)
- Joe C Udeochu
- Department of Anatomy University of California San Francisco San Francisco CA USA; Biomedical Sciences Graduate Program University of California San Francisco San Francisco CA USA
| | - Jeremy M Shea
- Department of Anatomy University of California San Francisco San Francisco CA USA
| | - Saul A Villeda
- Department of Anatomy University of California San Francisco San Francisco CA USA; Biomedical Sciences Graduate Program University of California San Francisco San Francisco CA USA; The Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research San Francisco CA USA
| |
Collapse
|
60
|
Zhang F, Nance E, Alnasser Y, Kannan R, Kannan S. Microglial migration and interactions with dendrimer nanoparticles are altered in the presence of neuroinflammation. J Neuroinflammation 2016; 13:65. [PMID: 27004516 PMCID: PMC4802843 DOI: 10.1186/s12974-016-0529-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/13/2016] [Indexed: 12/11/2022] Open
Abstract
Background Microglial cells have been implicated in neuroinflammation-mediated injury in the brain, including neurodevelopmental disorders such as cerebral palsy (CP) and autism. Pro-inflammatory activation of microglial cells results in the impairment of their neuroprotective functions, leading to an exaggerated, ongoing immune dysregulation that can persist long after the initial insult. We have previously shown that dendrimer-mediated delivery of an anti-inflammatory agent can attenuate inflammation in a rabbit model of maternal inflammation-induced CP and significantly improve the motor phenotype, due to the ability of the dendrimer to selectively localize in activated microglia. Methods To elucidate the interactions between dendrimers and microglia, we created an organotypic whole-hemisphere brain slice culture model from newborn rabbits with and without exposure to inflammation in utero. We then used this model to analyze the dynamics of microglial migration and their interactions with dendrimers in the presence of neuroinflammation. Results Microglial cells in animals with CP had an amoeboid morphology and impaired cell migration, demonstrated by decreased migration distance and velocity when compared to cells in healthy, age-matched controls. However, this decreased migration was associated with a greater, more rapid dendrimer uptake compared to microglial cells from healthy controls. Conclusions This study demonstrates that maternal intrauterine inflammation is associated with impaired microglial function and movement in the newborn brain. This microglial impairment may play a role in the development of ongoing brain injury and CP in the offspring. Increased uptake of dendrimers by the “impaired” microglia can be exploited to deliver drugs specifically to these cells and modulate their functions. Host tissue and target cell characteristics are important aspects to be considered in the design and evaluation of targeted dendrimer-based nanotherapeutics for improved and sustained efficacy. This ex vivo model also provides a rapid screening tool for evaluation of the effects of various therapies on microglial function. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0529-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Elizabeth Nance
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.,Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.,Present address: Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Yossef Alnasser
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Rangaramanujam Kannan
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.,Hugo Moser Research Center, Kennedy Krieger Institute, Baltimore, MD, 21205, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA. .,Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA. .,Hugo Moser Research Center, Kennedy Krieger Institute, Baltimore, MD, 21205, USA. .,Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
61
|
Cannon TD. How Schizophrenia Develops: Cognitive and Brain Mechanisms Underlying Onset of Psychosis. Trends Cogn Sci 2015; 19:744-756. [PMID: 26493362 DOI: 10.1016/j.tics.2015.09.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
Abstract
Identifying cognitive and neural mechanisms involved in the development of schizophrenia requires longitudinal observation of individuals prior to onset. Here recent studies of prodromal individuals who progress to full psychosis are briefly reviewed in relation to models of schizophrenia pathophysiology. Together, this body of work suggests that disruption in brain connectivity, driven primarily by a progressive reduction in dendritic spines on cortical pyramidal neurons, may represent a key triggering mechanism. The earliest disruptions appear to be in circuits involved in referencing experiences according to time, place, and agency, which may result in a failure to recognize particular cognitions as self-generated or to constrain interpretations of the meaning of events based on prior experiences, providing the scaffolding for faulty reality testing.
Collapse
Affiliation(s)
- Tyrone D Cannon
- Department of Psychology, Yale University, 2 Hillhouse Avenue, P.O. Box 208205, New Haven, CT 06520, USA.
| |
Collapse
|
62
|
Abstract
Neuroimaging studies have identified patterns of brain abnormalities in various stages of schizophrenia, but whether these abnormalities reflect primary factors associated with the causes of illness or secondary phenomena such as medications has been unclear. Recent work conducted within the prodromal risk paradigm suggests that progressive change in brain structure and function occurs around the time when clinically high-risk individuals transition into full-blown psychosis, effects that cannot be explained by exposure to medications or illness chronicity. This article reviews recent work bearing on the question of the timing of onset and course of brain changes, focusing on structural MRI, diffusion tensor imaging, and resting state connectivity MRI, in association with the onset and course of psychosis. We conclude with a consideration of potential mechanisms underlying progressive tissue changes during the prodromal phase of schizophrenia and implications for prevention.
Collapse
Affiliation(s)
- Yoonho Chung
- Deparment of Psychology, Yale University, New Haven CT
| | - Tyrone D. Cannon
- Deparment of Psychology, Yale University, New Haven CT
- Department of Psychiatry, Yale University, New Haven CT
| |
Collapse
|
63
|
Cannon TD, Chung Y, He G, Sun D, Jacobson A, van Erp TGM, McEwen S, Addington J, Bearden CE, Cadenhead K, Cornblatt B, Mathalon DH, McGlashan T, Perkins D, Jeffries C, Seidman LJ, Tsuang M, Walker E, Woods SW, Heinssen R. Progressive reduction in cortical thickness as psychosis develops: a multisite longitudinal neuroimaging study of youth at elevated clinical risk. Biol Psychiatry 2015; 77:147-57. [PMID: 25034946 PMCID: PMC4264996 DOI: 10.1016/j.biopsych.2014.05.023] [Citation(s) in RCA: 432] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/17/2014] [Accepted: 05/25/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Individuals at clinical high risk (CHR) who progress to fully psychotic symptoms have been observed to show a steeper rate of cortical gray matter reduction compared with individuals without symptomatic progression and with healthy control subjects. Whether such changes reflect processes associated with the pathophysiology of schizophrenia or exposure to antipsychotic drugs is unknown. METHODS In this multisite study, 274 CHR cases, including 35 individuals who converted to psychosis, and 135 healthy comparison subjects were scanned with magnetic resonance imaging at baseline, 12-month follow-up, or the point of conversion for the subjects who developed fully psychotic symptoms. RESULTS In a traveling subjects substudy, excellent reliability was observed for measures of cortical thickness and subcortical volumes. Controlling for multiple comparisons throughout the brain, CHR subjects who converted to psychosis showed a steeper rate of gray matter loss in the right superior frontal, middle frontal, and medial orbitofrontal cortical regions as well as a greater rate of expansion of the third ventricle compared with CHR subjects who did not convert to psychosis and healthy control subjects. Differential tissue loss was present in subjects who had not received antipsychotic medications during the interscan interval and was predicted by baseline levels of an aggregate measure of proinflammatory cytokines in plasma. CONCLUSIONS These findings demonstrate that the brain changes are not explained by exposure to antipsychotic drugs but likely play a role in psychosis pathophysiology. Given that the cortical changes were more pronounced in subjects with briefer durations of prodromal symptoms, contributing factors may predominantly play a role in acute-onset forms of psychosis.
Collapse
Affiliation(s)
- Tyrone D Cannon
- Departments of Psychology, Yale University, New Haven, Connecticut; Departments of Psychiatry, Yale University, New Haven, Connecticut.
| | - Yoonho Chung
- Departments of Psychology, Yale University, New Haven, Connecticut
| | - George He
- Departments of Psychology, Yale University, New Haven, Connecticut
| | - Daqiang Sun
- Semel Institute for Neuroscience and Human Behavior and Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Aron Jacobson
- Departments of Psychology, Yale University, New Haven, Connecticut
| | - Theo G M van Erp
- Department of Psychiatry, University of California, Irvine, Irvine, California
| | - Sarah McEwen
- Semel Institute for Neuroscience and Human Behavior and Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Jean Addington
- Department of Psychiatry (JA), University of Calgary, Calgary, Alberta, Canada
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior and Department of Psychology, University of California, Los Angeles, Los Angeles, California
| | - Kristin Cadenhead
- Department of Psychiatry, University of California, San Diego, San Diego, California
| | - Barbara Cornblatt
- Department of Psychiatry, Zucker Hillside Hospital, Glen Oaks, New York
| | - Daniel H Mathalon
- Department of Psychiatry, University of California, San Francisco, San Francisco, California
| | - Thomas McGlashan
- Departments of Psychiatry, Yale University, New Haven, Connecticut
| | - Diana Perkins
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Clark Jeffries
- Renaissance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Larry J Seidman
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ming Tsuang
- Department of Psychiatry, University of California, San Diego, San Diego, California
| | - Elaine Walker
- Department of Psychology, Emory University, Atlanta, Georgia
| | - Scott W Woods
- Departments of Psychiatry, Yale University, New Haven, Connecticut
| | - Robert Heinssen
- Division of Treatment and Prevention Research, National Institute of Mental Health, Rockville, Maryland
| |
Collapse
|
64
|
Aberrant production of tenascin-C in globoid cell leukodystrophy alters psychosine-induced microglial functions. J Neuropathol Exp Neurol 2014; 73:964-74. [PMID: 25192051 DOI: 10.1097/nen.0000000000000117] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Globoid cell leukodystrophy (GLD), or Krabbe disease, is a rare and often fatal demyelinating disease caused by mutations in the galactocerebrosidase (galc) gene that result in accumulation of galactosylsphingosine (psychosine). We recently reported that the extracellular matrix (ECM) protease, matrix metalloproteinase-3, is elevated in GLD and that it regulates psychosine-induced microglial activation. Here, we examined central nervous system ECM component expression in human GLD patients and in the twitcher mouse model of GLD using immunohistochemistry. The influence of ECM proteins on primary murine microglial responses to psychosine was evaluated using ECM proteins as substrates and analyzed by quantitative real-time polymerase chain reaction, immunocytochemistry, and ELISA. Functional analysis of microglial cytotoxicity was performed on oligodendrocytes in coculture, and cell death was measured by lactose dehydrogenase assay. Tenascin-C (TnC) was expressed at higher levels in human GLD and in twitcher mice versus controls. Microglial responses to psychosine were enhanced by TnC, as determined by an increase in globoid-like cell formation, matrix metalloproteinase-3 mRNA expression, and higher toxicity toward oligodendrocytes in culture. These findings were consistent with a shift toward the M1 microglial phenotype in TnC-grown microglia. Thus, elevated TnC expression in GLD modified microglial responses to psychosine. These data offer a novel perspective and enhance understanding of the microglial contribution to GLD pathogenesis.
Collapse
|
65
|
Martínez-Canabal A. Potential neuroprotective role of transforming growth factor β1 (TGFβ1) in the brain. Int J Neurosci 2014; 125:1-9. [PMID: 24628581 DOI: 10.3109/00207454.2014.903947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
TGFβ1 is a growth factor that is known to be expressed in most neurodegenerative diseases and after vascular accidents in the brain. TGFβ1 downregulates the activity of activated microglia and promotes astrogliosis. It also prevents cell death by a known mechanism dependant on astrocytes and the secretion of the plasminogen activator inhibitor 1 (PAI-1). This mechanism can provide light on what is the mechanism of action of TGFβ1 as a protective factor and it can provide the pharmacological principles in which this pathway could be used with therapeutic purposes. TGFβ1 is upregulated in most neurodegenerative diseases, however, its expression appears dramatically blocked in Huntington's disease, the fastest of those diseases in progress after the onset. This fact suggests that TGFβ1 slows down the neurodegenerative process, preventing tissue damage and neural apoptotic death. However, the exact details of TGFβ1 action are still unknown and the physiological roles on the diseases are still mysterious. Interestingly, all the data regarding the roles of TGFβ1 in health and disease have been also confirmed with the use of transgenic knockouts and TGFβ1 overexpressing mice. What possibly came as a surprise from the study of TGFβ1 overexpressing models is that combining its neuroprotective and antiproliferative effects, this cytokine generates a significant disruption in the hippocampal circuitry with its consequent learning and memory deficit.
Collapse
Affiliation(s)
- Alonso Martínez-Canabal
- Department of Molecular Neuropathology, Cell Physiology Institute (IFC), Department of Cell Biology, Faculty of Sciences, National Autonomous University of Mexico (UNAM). Ciudad Universitaria, Circuito exterior S/N, Coyoacan, 04510 Mexico D.F. Mexico
| |
Collapse
|
66
|
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: 167] [Impact Index Per Article: 16.7] [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
|
67
|
Abe C, Morita H. Drinking-induced bradyarrhythmias and cerebral injury in Dahl salt-sensitive rats with sinoaortic denervation. J Appl Physiol (1985) 2013; 115:1533-9. [PMID: 24072408 DOI: 10.1152/japplphysiol.00866.2013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have demonstrated that a drinking-induced pressor response was larger if the baroreflex did not operate, and the mean arterial pressure reached 163 mmHg in conscious rats with sinoaortic denervation (SAD). Thus we hypothesized that a drinking behavior became a cardiovascular risk factor if a basal arterial pressure was high. To clarify this, we analyzed the occurrence of arrhythmias and the accumulation of microglia in Dahl salt-sensitive rats (Dahl S) with SAD. We maintained Dahl S and Dahl salt-resistant rats (Dahl R) with a high-sodium diet for 5 weeks. After SAD surgery, we measured arterial pressure and electrocardiogram during water-drinking behavior in all rats. Furthermore, we measured tumor necrosis factor-α concentration in the cerebrospinal fluid (CSF) and microglial accumulations around the third and fourth ventricles in rats with programmed drinking at a rapid or slow rate for 7 days. Incidences of drinking-induced bradyarrhythmias and premature ventricular contractions (PVCs) were significantly larger in Dahl S than Dahl R rats. Both bradyarrhythmias and PVCs were completely abolished by atropine administration. Accumulations of microglia around the third ventricle and increases in TNF-α in the CSF were observed in rats that drank water at a rapid rate; these were not seen in rats that drank water slowly. In conclusion, both cardiovascular events and cerebral injury may be increased by drinking in Dahl S rats with SAD. These risks are reduced by modifying drinking behavior such as slowing the drinking rate.
Collapse
Affiliation(s)
- Chikara Abe
- Department of Physiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | | |
Collapse
|
68
|
Mindin is a critical mediator of ischemic brain injury in an experimental stroke model. Exp Neurol 2013; 247:506-16. [DOI: 10.1016/j.expneurol.2013.01.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 12/14/2012] [Accepted: 01/18/2013] [Indexed: 11/23/2022]
|
69
|
Stolzing A, Sethe S, Grune T. Chronically active: activation of microglial proteolysis in ageing and neurodegeneration. Redox Rep 2013; 10:207-13. [PMID: 16259788 DOI: 10.1179/135100005x70198] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
One of the microglial cell functions is the removal of modified extracellular proteins in the brain. The connection between protein oxidation, proteolysis, and microglial activation is the topic of this review. The effect of various activation agents on microglial cells with regard to changes in substrate uptake, proteolytic capacity and degradation efficiency of different types of oxidized protein materials is reviewed. It is shown that different activation stimuli initiate substrate-specific modulation for uptake and proteolysis, influencing an array of factors including receptor expression, lysosomal pH, and proteasome subunit composition. Age-related alterations in activation and proteolytic capacity in microglial cells are also discussed. In ageing, proteolytic effectiveness is diminished, while microglial cells are chronically activated and lose the oxidative burst ability, possibly supporting a 'vicious circle' of macrophage-induced neurodegeneration.
Collapse
Affiliation(s)
- Alexandra Stolzing
- Centre for Biomaterials and Tissue Engineering, Sheffield University, UK
| | | | | |
Collapse
|
70
|
Welser-Alves JV, Milner R. Microglia are the major source of TNF-α and TGF-β1 in postnatal glial cultures; regulation by cytokines, lipopolysaccharide, and vitronectin. Neurochem Int 2013; 63:47-53. [PMID: 23619393 DOI: 10.1016/j.neuint.2013.04.007] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 03/22/2013] [Accepted: 04/15/2013] [Indexed: 12/15/2022]
Abstract
Damage to the central nervous system (CNS) leads to increased production of TNF-α and TGF-β1 cytokines that have pro- or anti-inflammatory actions, respectively. To define whether astrocytes or microglia express these cytokines, prior studies have used mixed glial cultures (MGC) to represent astrocytes, thought these results are inevitably complicated by the presence of contaminating microglia within MGC. To clarify the cellular source of these cytokines, here we employed a recently described method of preparing microglia-free astrocyte cultures, in which neural stem cells (NSC) are differentiated into astrocytes. Using ELISA to quantify cytokine production in three types of glial culture: MGC, pure microglia or pure astrocytes, this showed that microglia but not astrocytes, produce TNF-α, and that this expression is increased by LPS, IFN-γ, and to a lesser extent by vitronectin, but decreased by TGF-β1. In contrast, TGF-β1 was produced by microglia and astrocytes, though at 10-fold higher levels by microglia. TGF-β1 expression in microglia was increased by vitronectin and to a lesser extent by TNF-α and LPS, but astrocyte TGF-β1 expression was not regulated by any factor tested. In summary, our data reveal that microglia, not astrocytes are the major source of TNF-α and TGF-β1 in postnatal glial cultures, and that microglial production of these antagonistic cytokines is tightly regulated by cytokines, LPS, and vitronectin.
Collapse
Affiliation(s)
- Jennifer V Welser-Alves
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
71
|
Kim H, Ahn M, Choi S, Kim M, Sim KB, Kim J, Moon C, Shin T. Potential role of fibronectin in microglia/macrophage activation following cryoinjury in the rat brain: An immunohistochemical study. Brain Res 2013; 1502:11-9. [DOI: 10.1016/j.brainres.2013.01.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 12/18/2012] [Accepted: 01/27/2013] [Indexed: 01/17/2023]
|
72
|
Tigges U, Boroujerdi A, Welser-Alves JV, Milner R. TNF-α promotes cerebral pericyte remodeling in vitro, via a switch from α1 to α2 integrins. J Neuroinflammation 2013; 10:33. [PMID: 23448258 PMCID: PMC3616978 DOI: 10.1186/1742-2094-10-33] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 02/15/2013] [Indexed: 01/07/2023] Open
Abstract
Background There is increasing evidence to suggest that pericytes play a crucial role in regulating the remodeling state of blood vessels. As cerebral pericytes are embedded within the extracellular matrix (ECM) of the vascular basal lamina, it is important to understand how individual ECM components influence pericyte remodeling behavior, and how cytokines regulate these events. Methods The influence of different vascular ECM substrates on cerebral pericyte behavior was examined in assays of cell adhesion, migration, and proliferation. Pericyte expression of integrin receptors was examined by flow cytometry. The influence of cytokines on pericyte functions and integrin expression was also examined, and the role of specific integrins in mediating these effects was defined by function-blocking antibodies. Expression of pericyte integrins within remodeling cerebral blood vessels was analyzed using dual immunofluorescence (IF) of brain sections derived from the animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Results Fibronectin and collagen I promoted pericyte proliferation and migration, but heparan sulfate proteoglycan (HSPG) had an inhibitory influence on pericyte behavior. Flow cytometry showed that cerebral pericytes express high levels of α5 integrin, and lower levels of α1, α2, and α6 integrins. The pro-inflammatory cytokine tumor necrosis factor (TNF)-α strongly promoted pericyte proliferation and migration, and concomitantly induced a switch in pericyte integrins, from α1 to α2 integrin, the opposite to the switch seen when pericytes differentiated. Inhibition studies showed that α2 integrin mediates pericyte adhesion to collagens, and significantly, function blockade of α2 integrin abrogated the pro-modeling influence of TNF-α. Dual-IF on brain tissue with the pericyte marker NG2 showed that while α1 integrin was expressed by pericytes in both stable and remodeling vessels, pericyte expression of α2 integrin was strongly induced in remodeling vessels in EAE brain. Conclusions Our results suggest a model in which ECM constituents exert an important influence on pericyte remodeling status. In this model, HSPG restricts pericyte remodeling in stable vessels, but during inflammation, TNF-α triggers a switch in pericyte integrins from α1 to α2, thereby stimulating pericyte proliferation and migration on collagen. These results thus define a fundamental molecular mechanism in which TNF-α stimulates pericyte remodeling in an α2 integrin-dependent manner.
Collapse
Affiliation(s)
- Ulrich Tigges
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla CA, 92037, USA
| | | | | | | |
Collapse
|
73
|
Eyo UB, Dailey ME. Microglia: key elements in neural development, plasticity, and pathology. J Neuroimmune Pharmacol 2013; 8:494-509. [PMID: 23354784 DOI: 10.1007/s11481-013-9434-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 01/14/2013] [Indexed: 12/31/2022]
Abstract
A century after Cajal identified a "third element" of the nervous system, many issues have been clarified about the identity and function of one of its major components, the microglia. Here, we review recent findings by microgliologists, highlighting results from imaging studies that are helping provide new views of microglial behavior and function. In vivo imaging in the intact adult rodent CNS has revolutionized our understanding of microglial behaviors in situ and has raised speculation about their function in the uninjured adult brain. Imaging studies in ex vivo mammalian tissue preparations and in intact model organisms including zebrafish are providing insights into microglial behaviors during brain development. These data suggest that microglia play important developmental roles in synapse remodeling, developmental apoptosis, phagocytic clearance, and angiogenesis. Because microglia also contribute to pathology, including neurodevelopmental and neurobehavioral disorders, ischemic injury, and neuropathic pain, promising new results raise the possibility of leveraging microglia for therapeutic roles. Finally, exciting recent work is addressing unanswered questions regarding the nature of microglial-neuronal communication. While it is now apparent that microglia play diverse roles in neural development, behavior, and pathology, future research using neuroimaging techniques will be essential to more fully exploit these intriguing cellular targets for effective therapeutic intervention applied to a variety of conditions.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
74
|
Yao H, Duan M, Yang L, Buch S. Nonmuscle myosin light-chain kinase mediates microglial migration induced by HIV Tat: involvement of β1 integrins. FASEB J 2013; 27:1532-48. [PMID: 23292072 DOI: 10.1096/fj.12-219600] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
One of the hallmark features of HIV-associated neurological disease is increased activation and migration of microglia. HIV transactivator of transcription (Tat) is released from infected cells and has the ability to recruit microglia. The purpose of this study was to investigate molecular mechanisms by which recombinant Tat₁₋₇₂, but not heated-inactive Tat₁₋₇₂,induces migration of rat primary microglia. Using primary microglia in Boyden chambers, we demonstrated the role of nonmuscle myosin light-chain kinase (nmMYLK) in Tat₁₋₇₂ (14.4 nM)-mediated increased microglial migration (up to 171.85%). These findings were validated using microglia isolated from wild-type (WT) or nmMYLK(-/-) mice in Dunn chamber assays. Tat₁₋₇₂-mediated activation of nmMYLK resulted in "inside-out" activation of β1 integrin, followed by "outside-in" activation of c-Src, Pyk2, and Cdc42-GTP (using G-LISA in primary and nmMYLK(-/-) microglia) and, subsequently, actin polymerization (flow cytometry and Western blot assays). In vivo corroboration of these findings demonstrated decreased migration of nmMYLK(-/-) microglia (2 × 10(5) cells transplanted into corpus callosum) compared with WT microglia toward microinjected Tat₁₋₇₂ (2 μg/mouse) in hippocampus. Up-regulation of nmMYLK in microglia was also detected in sections of basal ganglia from humans with HIV-encephalitis compared with uninfected controls. nmMYLK is thus critical for eliciting microglial migration during the innate immune response.
Collapse
Affiliation(s)
- Honghong Yao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198-5880, USA
| | | | | | | |
Collapse
|
75
|
Li J, Yang JY, Yao XC, Xue X, Zhang QC, Wang XX, Ding LL, Wu CF. Oligomeric Aβ-induced microglial activation is possibly mediated by NADPH oxidase. Neurochem Res 2012; 38:443-52. [PMID: 23229789 DOI: 10.1007/s11064-012-0939-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 11/07/2012] [Accepted: 11/23/2012] [Indexed: 12/15/2022]
Abstract
Recent studies have shown that oligomeric amyloid-β (oAβ) peptide can potentially activate microglia in addition to inducing more potent neurotoxicity compared with fibrillar Aβ (fAβ); however, its mechanisms of action remain unclear. This study was designed to investigate the possible mechanisms involved in the microglial activation induced by oAβ in BV-2 microglial cells. The results showed that oAβ induced activated properties of microglia, including higher proliferative capacity as well as increased production of reactive oxygen species, nitric oxide (NO), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β). NADPH oxidase inhibitors [diphenylene iodonium (DPI) and apocynin (4-hydroxy-3-methoxy-acetophenone)] prevented the microglial activation induced by oAβ, suggesting that NADPH oxidase activation was involved in microglial activation. In addition, TNF-α and IL-1β, which are massively released by activated microglia, significantly induced the activation of microglia, thereby resulting in the production of NO and proliferation of microglia, respectively. These effects could be inhibited by diphenylene iodonium and apocynin, indicating a self-cycle regulated by NADPH oxidase in microglial activation in response to oAβ. In conclusion, microglial activation induced by oAβ is possibly mediated by NADPH oxidase, suggesting that oAβ, which is normally considered a neurotoxin, may also lead to indirect neuronal damage through the pro-inflammation activation of microglia in Alzheimer's disease and that NADPH oxidase could be a potential target to prevent oAβ-induced inflammatory neurodegeneration.
Collapse
Affiliation(s)
- Juan Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Box 31, Shenyang 110016, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
76
|
The CCL2 synthesis inhibitor bindarit targets cells of the neurovascular unit, and suppresses experimental autoimmune encephalomyelitis. J Neuroinflammation 2012; 9:171. [PMID: 22788993 PMCID: PMC3488971 DOI: 10.1186/1742-2094-9-171] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/12/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Production of the chemokine CCL2 by cells of the neurovascular unit (NVU) drives critical aspects of neuroinflammation. Suppression of CCL2 therefore holds promise in treating neuroinflammatory disease. Accordingly, we sought to determine if the compound bindarit, which inhibits CCL2 synthesis, could repress the three NVU sources of CCL2 most commonly reported in neuroinflammation--astrocytes, microglia and brain microvascular endothelial cells (BMEC)--as well as modify the clinical course of neuroinflammatory disease. METHODS The effect of bindarit on CCL2 expression by cultured murine astrocytes, microglia and BMEC was examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Bindarit action on mouse brain and spinal cord in vivo was similarly investigated by qRT-PCR following LPS injection in mice. And to further gauge the potential remedial effects of bindarit on neuroinflammatory disease, its impact on the clinical course of experimental autoimmune encephalomyelitis (EAE) in mice was also explored. RESULTS Bindarit repressed CCL2 expression by all three cultured cells, and antagonized upregulated expression of CCL2 in both brain and spinal cord in vivo following LPS administration. Bindarit also significantly modified the course and severity of clinical EAE, diminished the incidence and onset of disease, and evidenced signs of disease reversal. CONCLUSION Bindarit was effective in suppressing CCL2 expression by cultured NVU cells as well as brain and spinal cord tissue in vivo. It further modulated the course of clinical EAE in both preventative and therapeutic ways. Collectively, these results suggest that bindarit might prove an effective treatment for neuroinflammatory disease.
Collapse
|
77
|
Conant K, Lim ST, Randall B, Maguire-Zeiss KA. Matrix metalloproteinase dependent cleavage of cell adhesion molecules in the pathogenesis of CNS dysfunction with HIV and methamphetamine. Curr HIV Res 2012; 10:384-91. [PMID: 22591362 PMCID: PMC6035363 DOI: 10.2174/157016212802138733] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 01/18/2012] [Accepted: 01/25/2012] [Indexed: 01/15/2023]
Abstract
Physiologically appropriate levels of matrix metalloproteinases (MMPs) are likely important to varied aspects of CNS function. In particular, these enzymes may contribute to neuronal activity dependent synaptic plasticity and to cell mobility in processes including stem cell migration and immune surveillance. Levels of MMPs may, however, be substantially increased in the setting of HIV infection with methamphetamine abuse. Elevated MMP levels might in turn influence integrity of the blood brain barrier, as has been demonstrated in published work. Herein we suggest that elevated levels of MMPs can also contribute to microglial activation as well as neuronal and synaptic injury through a mechanism that involves cleavage of specific cell and synaptic adhesion molecules.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Research Building EP-16, 3970 Reservoir Rd, Washington, DC 20007, USA.
| | | | | | | |
Collapse
|
78
|
Yeh WL, Lu DY, Liou HC, Fu WM. A forward loop between glioma and microglia: glioma-derived extracellular matrix-activated microglia secrete IL-18 to enhance the migration of glioma cells. J Cell Physiol 2012; 227:558-68. [PMID: 21442623 DOI: 10.1002/jcp.22746] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The mediators and cellular effectors of inflammation are important constituents of the local environment of tumors. In some occasions, oncogenic changes induce an inflammatory microenvironment that promotes the progression of tumors. In gliomas, the presence of microglia may represent tumor-related inflammation and microglia activation, and subsequent inflammatory responses may influence tumor growth and metastasis. Here, we found that C6 glioma--but not primary astrocyte-derived extracellular matrix (ECM) could activate microglia, including primary microglia and BV-2 cell line, and activated microglia-secreted interleukin (IL)-18, a potent inflammatory cytokine of the IL-1 family, to promote C6 migration. In addition, by coating purified ECM components, it was found that secretion of IL-18 by activated microglia was enhanced when microglia encountered with fibronectin and vitronectin. Furthermore, IL-18-induced C6 migration and microfilament disassembly were antagonized by iNOS inhibitor, guanylate cyclase inhibitor, and protein kinase G inhibitor. Taken together, these results indicate that IL-18 secreted by microglia, which was activated by C6 glioma-derived ECM, enhanced migration of C6 glioma through NO/cGMP pathway.
Collapse
Affiliation(s)
- Wei-Lan Yeh
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
79
|
Colombatti A, Spessotto P, Doliana R, Mongiat M, Bressan GM, Esposito G. The EMILIN/Multimerin family. Front Immunol 2012; 2:93. [PMID: 22566882 PMCID: PMC3342094 DOI: 10.3389/fimmu.2011.00093] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/21/2011] [Indexed: 01/12/2023] Open
Abstract
Elastin microfibrillar interface proteins (EMILINs) and Multimerins (EMILIN1, EMILIN2, Multimerin1, and Multimerin2) constitute a four member family that in addition to the shared C-terminus gC1q domain typical of the gC1q/TNF superfamily members contain a N-terminus unique cysteine-rich EMI domain. These glycoproteins are homotrimeric and assemble into high molecular weight multimers. They are predominantly expressed in the extracellular matrix and contribute to several cellular functions in part associated with the gC1q domain and in part not yet assigned nor linked to other specific regions of the sequence. Among the latter is the control of arterial blood pressure, the inhibition of Bacillus anthracis cell cytotoxicity, the promotion of cell death, the proangiogenic function, and a role in platelet hemostasis. The focus of this review is to highlight the multiplicity of functions and domains of the EMILIN/Multimerin family with a particular emphasis on the regulatory role played by the ligand-receptor interactions of the gC1q domain. EMILIN1 is the most extensively studied member both from the structural and functional point of view. The structure of the gC1q of EMILIN1 solved by NMR highlights unique characteristics compared to other gC1q domains: it shows a marked decrease of the contact surface of the trimeric assembly and while conserving the jelly-roll topology with two β-sheets of antiparallel strands it presents a nine-stranded β-sandwich fold instead of the usual 10-stranded fold. This is likely due to the insertion of nine residues that disrupt the ordered strand organization and forma a highly dynamic protruding loop. In this loop the residue E933 is the site of interaction between gC1q and the α4β1 and α9β1 integrins, and contrary to integrin occupancy that usually upregulates cell growth, when gC1q is ligated by the integrin the cells reduce their proliferative activity.
Collapse
Affiliation(s)
- Alfonso Colombatti
- Experimental Oncology 2, Centro di Riferimento Oncologico, Istituto di Ricerca e Cura a Carattere Scientifico Aviano, Italy.
| | | | | | | | | | | |
Collapse
|
80
|
Use of astrocyte-microglial cocultures to examine the regulatory influence of astrocytes on microglial activation. Methods Mol Biol 2012; 814:367-80. [PMID: 22144319 DOI: 10.1007/978-1-61779-452-0_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Microglia are the principal immune effector cells of the central nervous system (CNS). Under normal conditions, they occupy a quiescent surveillance phenotype, but following stimulation by microorganisms or inflammatory cytokines, microglia transform into highly activated migratory, phagocytic cells producing inflammatory cytokines and chemokines. Significantly, several studies have demonstrated that astrocytes attenuate microglial activation, reducing microglial adhesion, production of interleukin-12 (IL-12) and reactive oxygen species (ROS), and expression of inducible nitric oxide synthase (iNOS). In this chapter, we describe an astrocyte-microglia coculture system that can be used to investigate interactions between these two cell types. We also describe a flow cytometry approach to quantify microglial activation state, as assessed by microglial expression of cellular activation markers, including MHC class I and the Mac-1 and α4 integrins.
Collapse
|
81
|
Bradaric BD, Patel A, Schneider JA, Carvey PM, Hendey B. Evidence for angiogenesis in Parkinson's disease, incidental Lewy body disease, and progressive supranuclear palsy. J Neural Transm (Vienna) 2012; 119:59-71. [PMID: 21748523 PMCID: PMC3352316 DOI: 10.1007/s00702-011-0684-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 06/29/2011] [Indexed: 12/21/2022]
Abstract
Angiogenesis has not been extensively studied in Parkinson's disease (PD) despite being associated with other neurodegenerative disorders. Post-mortem human brain tissues were obtained from subjects with pathologically confirmed Parkinson's disease (PD) and progressive supranuclear palsy (PSP), a rapidly progressing Parkinsonian-like disorder. Tissues were also obtained from subjects with incidental Lewy body disease (iLBD) who had Lewy bodies in the substantia nigra pars compacta (SN(pc)) but had not been diagnosed with PD, and age-matched controls without Lewy body pathology. The SNpc, putamen, locus ceruleus (LC) and midfrontal cortex were examined for integrin αvβ3, a marker for angiogenesis, along with vessel number and activated microglia. All parkinsonian syndromes had greater αvβ3 in the LC and the SN(pc), while only PD and PSP subjects had elevated αvβ3 in the putamen compared to controls. PD and PSP subjects also had increases in microglia number and activation in the SN(pc) suggesting a link between inflammation and clinical disease. Microglia activation in iLBD subjects was limited to the LC, an area involved at an early stage of PD. Likewise, iLBD subjects did not differ from controls in αvβ3 staining in the putamen, a late area of involvement in PD. The presence of αvβ3 reactive vessels in PD and its syndromes is indicative of newly created vessels that have not likely developed the restrictive properties of the blood brain barrier. Such angiogenic vessels could contribute to neuroinflammation by failing to protect the parenchyma from peripheral immune cells and inflammatory or toxic factors in the peripheral circulation.
Collapse
Affiliation(s)
| | - Aditiben Patel
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University, Chicago, IL 60612, USA
- Department of Pathology, Rush University, Chicago, IL 60612, USA
| | - Paul M. Carvey
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University, Chicago, IL 60612, USA
| | - Bill Hendey
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
| |
Collapse
|
82
|
Welser-Alves JV, Boroujerdi A, Tigges U, Milner R. Microglia use multiple mechanisms to mediate interactions with vitronectin; non-essential roles for the highly-expressed αvβ3 and αvβ5 integrins. J Neuroinflammation 2011; 8:157. [PMID: 22074485 PMCID: PMC3239846 DOI: 10.1186/1742-2094-8-157] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 11/10/2011] [Indexed: 11/10/2022] Open
Abstract
Background As the primary resident immune cells, microglia play a central role in regulating inflammatory processes in the CNS. The extracellular matrix (ECM) protein vitronectin promotes microglial activation, switching microglia into an activated phenotype. We have shown previously that microglia express two vitronectin receptors, αvβ3 and αvβ5 integrins. As these integrins have well-defined roles in activation and phagocytic processes in other cell types, the purpose of the current study was to investigate the contribution of these two integrins in microglial activation. Methods Microglial cells were prepared from wild-type, β3 integrin knockout (KO), β5 integrin KO or β3/β5 integrin DKO mice, and their interactions and activation responses to vitronectin examined in a battery of assays, including adhesion, expression of activation markers, MMP-9 expression, and phagocytosis. Expression of other αv integrins was examined by flow cytometry and immunoprecipitation. Results Surprisingly, when cultured on vitronectin, microglia from the different knockout strains showed no obvious defects in adhesion, activation marker expression, MMP-9 induction, or phagocytosis of vitronectin-coated beads. To investigate the reason for this lack of effect, we examined the expression of other αv integrins. Flow cytometry showed that β3/β5 integrin DKO microglia expressed residual αv integrin at the cell surface, and immunoprecipitation confirmed this finding by revealing the presence of low levels of the αvβ1 and αvβ8 integrins. β1 integrin blockade had no impact on adhesion of β3/β5 integrin DKO microglia to vitronectin, suggesting that in addition to αvβ1, αvβ3, and αvβ5, αvβ8 also serves as a functional vitronectin receptor on microglia. Conclusions Taken together, this demonstrates that the αvβ3 and αvβ5 integrins are not essential for mediating microglial activation responses to vitronectin, but that microglia use multiple redundant receptors to mediate interactions with this ECM protein.
Collapse
Affiliation(s)
- Jennifer V Welser-Alves
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
83
|
Patel A, Toia GV, Colletta K, Bradaric BD, Carvey PM, Hendey B. An angiogenic inhibitor, cyclic RGDfV, attenuates MPTP-induced dopamine neuron toxicity. Exp Neurol 2011; 231:160-70. [PMID: 21703263 DOI: 10.1016/j.expneurol.2011.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 05/23/2011] [Accepted: 06/07/2011] [Indexed: 12/17/2022]
Abstract
We previously demonstrated that several dopamine (DA) neurotoxins produced punctate areas of FITC-labeled albumin (FITC-LA) leakage in the substantia nigra and striatum suggesting blood brain barrier (BBB) dysfunction. Further, this leakage was co-localized with αvβ3 integrin up-regulation, a marker for angiogenesis. This suggested that the FITC-LA leakage might have been a result of angiogenesis. To assess the possible role of angiogenesis in DA neuron loss, we treated mice with 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP) and on the following day treated with cyRGDfV, a cyclic peptide that binds to integrin αvβ3 and prevents angiogenesis. Post-treatment for 3 days (b.i.d.) with cyRGDfV blocked the MPTP-induced upregulation of integrin β3 immunoreactivity (a marker for angiogenesis), leakage of FITC-LA into brain parenchyma (a marker for BBB disruption) as well as the down regulation of Zona Occludin-1 (ZO-1; a marker for tight junction integrity). In addition, cyRGDfV also completely prevented tyrosine hydroxylase immunoreactive cell loss (a marker for DA neurons) and markedly attenuated the up-regulation of activated microglia (Iba1 cell counts and morphology). These data suggest that cyRGDfV, and perhaps other anti-angiogenic drugs, are neuroprotective following acute MPTP treatment and may suggest that compensatory angiogenesis and BBB dysfunction may contribute to inflammation and DA neuron loss.
Collapse
Affiliation(s)
- Aditiben Patel
- Department of Pharmacology, Rush University, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
84
|
Rodríguez-González R, Sobrino T, Rodríguez-Yáñez M, Millán M, Brea D, Miranda E, Moldes O, Pérez J, Lomas DA, Leira R, Dávalos A, Castillo J. Association between neuroserpin and molecular markers of brain damage in patients with acute ischemic stroke. J Transl Med 2011; 9:58. [PMID: 21569344 PMCID: PMC3113955 DOI: 10.1186/1479-5876-9-58] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 05/11/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroserpin has shown neuroprotective effects in animal models of cerebral ischemia and has been associated with functional outcome after ischemic stroke. Our aim was to study whether neuroserpin serum levels could be associated to biomarkers of excitotoxicity, inflammation and blood brain barrier disruption. METHODS We prospectively included 129 patients with ischemic stroke (58.1% male; mean age, 72.4 ± 9.6 years) not treated with tPA within 12 hours (h) of symptoms onset (mean time, 4.7 ± 2.1 h). Poor functional outcome at 3 months was considered as a modified Rankin scale score >2. Serum levels of neuroserpin, Interleukin 6 (IL-6), Intercellular adhesion molecule-1 (ICAM-1), active Matrix metalloproteinase 9 (MMP-9), and cellular fibronectin (cFn) (determined by ELISA) and glutamate (determined by HPLC) were measured on admission, 24 and 72 h. The main variable was considered the decrease of neuroserpin levels within the first 24 h. ROC analysis was used to select the best predictive value for neuroserpin to predict poor functional outcome due to a lack of linearity. RESULTS The decrease of neuroserpin levels within the first 24 h was negatively correlated with serum levels at 24 hours of glutamate (r = -0.642), IL-6 (r = -0.678), ICAM-1 (r = -0.345), MMP-9 (r = -0.554) and cFn (r = -0.703) (all P < 0.0001). In the multivariate analysis, serum levels of glutamate (OR, 1.04; CI95%, 1.01-1.06, p = 0.001); IL-6 (OR, 1.4; CI95%, 1.1-1.7, p = 0.001); and cFn (OR, 1.3; CI95%, 1.1-1.6, p = 0.002) were independently associated with a decrease of neuroserpin levels <70 ng/mL at 24 h after adjusting for confounding factors. CONCLUSIONS These findings suggest that neuroprotective properties of neuroserpin may be related to the inhibition of excitotoxicity, inflammation, as well as blood brain barrier disruption that occur after acute ischemic stroke.
Collapse
Affiliation(s)
- Raquel Rodríguez-González
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Rodríguez-Yáñez
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mónica Millán
- Department of Neurosciences, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - David Brea
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Elena Miranda
- University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Octavio Moldes
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan Pérez
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Facultad de Ciencias, Campus de Teatinos, Málaga, Spain
| | - David A Lomas
- University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Rogelio Leira
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Antoni Dávalos
- Department of Neurosciences, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - José Castillo
- Clinical Neuroscience Research Laboratory, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
85
|
Neher JJ, Neniskyte U, Zhao JW, Bal-Price A, Tolkovsky AM, Brown GC. Inhibition of microglial phagocytosis is sufficient to prevent inflammatory neuronal death. THE JOURNAL OF IMMUNOLOGY 2011; 186:4973-83. [PMID: 21402900 DOI: 10.4049/jimmunol.1003600] [Citation(s) in RCA: 288] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is well-known that dead and dying neurons are quickly removed through phagocytosis by the brain's macrophages, the microglia. Therefore, neuronal loss during brain inflammation has always been assumed to be due to phagocytosis of neurons subsequent to their apoptotic or necrotic death. However, we report in this article that under inflammatory conditions in primary rat cultures of neurons and glia, phagocytosis actively induces neuronal death. Specifically, two inflammatory bacterial ligands, lipoteichoic acid or LPS (agonists of glial TLR2 and TLR4, respectively), stimulated microglial proliferation, phagocytic activity, and engulfment of ∼30% of neurons within 3 d. Phagocytosis of neurons was dependent on the microglial release of soluble mediators (and peroxynitrite in particular), which induced neuronal exposure of the eat-me signal phosphatidylserine (PS). Surprisingly, however, eat-me signaling was reversible, so that blocking any step in a phagocytic pathway consisting of PS exposure, the PS-binding protein milk fat globule epidermal growth factor-8, and its microglial vitronectin receptor was sufficient to rescue up to 90% of neurons without reducing inflammation. Hence, our data indicate a novel form of inflammatory neurodegeneration, where inflammation can cause eat-me signal exposure by otherwise viable neurons, leading to their death through phagocytosis. Thus, blocking phagocytosis may prevent some forms of inflammatory neurodegeneration, and therefore might be beneficial during brain infection, trauma, ischemia, neurodegeneration, and aging.
Collapse
Affiliation(s)
- Jonas J Neher
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom.
| | | | | | | | | | | |
Collapse
|
86
|
Perrin RJ, Craig-Schapiro R, Malone JP, Shah AR, Gilmore P, Davis AE, Roe CM, Peskind ER, Li G, Galasko DR, Clark CM, Quinn JF, Kaye JA, Morris JC, Holtzman DM, Townsend RR, Fagan AM. Identification and validation of novel cerebrospinal fluid biomarkers for staging early Alzheimer's disease. PLoS One 2011; 6:e16032. [PMID: 21264269 PMCID: PMC3020224 DOI: 10.1371/journal.pone.0016032] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 12/03/2010] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ideally, disease modifying therapies for Alzheimer disease (AD) will be applied during the 'preclinical' stage (pathology present with cognition intact) before severe neuronal damage occurs, or upon recognizing very mild cognitive impairment. Developing and judiciously administering such therapies will require biomarker panels to identify early AD pathology, classify disease stage, monitor pathological progression, and predict cognitive decline. To discover such biomarkers, we measured AD-associated changes in the cerebrospinal fluid (CSF) proteome. METHODS AND FINDINGS CSF samples from individuals with mild AD (Clinical Dementia Rating [CDR] 1) (n = 24) and cognitively normal controls (CDR 0) (n = 24) were subjected to two-dimensional difference-in-gel electrophoresis. Within 119 differentially-abundant gel features, mass spectrometry (LC-MS/MS) identified 47 proteins. For validation, eleven proteins were re-evaluated by enzyme-linked immunosorbent assays (ELISA). Six of these assays (NrCAM, YKL-40, chromogranin A, carnosinase I, transthyretin, cystatin C) distinguished CDR 1 and CDR 0 groups and were subsequently applied (with tau, p-tau181 and Aβ42 ELISAs) to a larger independent cohort (n = 292) that included individuals with very mild dementia (CDR 0.5). Receiver-operating characteristic curve analyses using stepwise logistic regression yielded optimal biomarker combinations to distinguish CDR 0 from CDR>0 (tau, YKL-40, NrCAM) and CDR 1 from CDR<1 (tau, chromogranin A, carnosinase I) with areas under the curve of 0.90 (0.85-0.94 95% confidence interval [CI]) and 0.88 (0.81-0.94 CI), respectively. CONCLUSIONS Four novel CSF biomarkers for AD (NrCAM, YKL-40, chromogranin A, carnosinase I) can improve the diagnostic accuracy of Aβ42 and tau. Together, these six markers describe six clinicopathological stages from cognitive normalcy to mild dementia, including stages defined by increased risk of cognitive decline. Such a panel might improve clinical trial efficiency by guiding subject enrollment and monitoring disease progression. Further studies will be required to validate this panel and evaluate its potential for distinguishing AD from other dementing conditions.
Collapse
Affiliation(s)
- Richard J Perrin
- Division of Neuropathology, Washington University School of Medicine, St. Louis, Missouri, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Achyuta AKH, Stephens KD, Pryce Lewis HG, Murthy SK. Mitigation of reactive human cell adhesion on poly(dimethylsiloxane) by immobilized trypsin. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2010; 26:4160-7. [PMID: 20214394 PMCID: PMC2924170 DOI: 10.1021/la903441u] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Occlusion or blockage of silicone shunts utilized in the treatment of hydrocephalus is a major challenge that is currently addressed by multiple shunt replacements. Shunt occlusion is caused by the adhesion and proliferation of reactive cells, such as glial and vascular cells, into the lumen of the catheter and on valve components. This in vitro study describes how the adhesive behavior of four human cell types on poly(dimethylsiloxane) (PDMS) surfaces can be suppressed by functionalization with trypsin, a proteolytic enzyme. The covalently conjugated trypsin retained its proteolytic activity and acted in a dose-dependent manner. Trypsin-modified PDMS surfaces supported significantly lower adhesion of normal human astrocytes, human microglia, human dermal fibroblasts, and human umbilical vein endothelial cells compared to unmodified PDMS surfaces (p < 0.0001). Immunofluorescence imaging of cellular fibronectin and quantitative adsorption experiments with serum components indicated that the PDMS surfaces immobilized with trypsin inhibited surface remodeling by all cell types and resisted protein adsorption. The impact of this work lies in the recognition that the well-known proteolytic characteristics of trypsin can be harnessed by covalent surface immobilization to suppress cell adhesion and protein adsorption.
Collapse
Affiliation(s)
- Anil Kumar H. Achyuta
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 342 SN, Boston, Massachusetts 02115, USA
| | - Kyle D. Stephens
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 342 SN, Boston, Massachusetts 02115, USA
| | | | - Shashi K. Murthy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 342 SN, Boston, Massachusetts 02115, USA
| |
Collapse
|
88
|
Ribes S, Ebert S, Regen T, Czesnik D, Scheffel J, Zeug A, Bunkowski S, Eiffert H, Hanisch UK, Hammerschmidt S, Nau R. Fibronectin stimulates Escherichia coli phagocytosis by microglial cells. Glia 2010; 58:367-76. [PMID: 19780198 DOI: 10.1002/glia.20929] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Microglia express Toll-like receptors (TLRs) that recognize invading pathogens as well as endogenous proteins such as fibronectin under nonphysiological conditions. Here, we demonstrated that fibronectin stimulates murine microglia in culture in a dose-dependent manner: microglial cells secreted proinflammatory cytokines and chemokines and increased phagocytosis of Escherichia coli DH5alpha and E. coli K1 strains. Low levels of fibronectin exerted a synergistic effect on the release of proinflammatory compounds by microglia co-stimulated with agonists for TLR1/2 (Pam(3)CSK(4)) or TLR9 (CpG DNA), but not in combination with the TLR4 agonist lipopolysaccharide (LPS). Phagocytosis of bacterial strains was moderately enhanced when microglia was co-stimulated with high concentrations of fibronectin and one pathogen-derived TLR agonist. In conclusion, fibronectin increased proinflammatory and phagocytotic functions in microglia and partially synergized with microbial TLR agonists.
Collapse
Affiliation(s)
- Sandra Ribes
- Department of Neurology, University of Göttingen, Robert-Koch-Strasse 40, Göttingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Abstract
Microvessels and neurons respond rapidly and simultaneously in focal regions of ischaemic injury in such a way as to suggest that the responses could be coordinated. The ability of neurons to modulate cerebral blood flow in regions of activation results from neurovascular coupling. But little is known about the microvessel-to-neuron direction of the relationship. The presence and participation of intervening glial cells implies the association of microvessels, glia, and neurons in a 'neurovascular unit'. The interdependent functions of the cellular and matrix components of this theoretical unit have not been rigorously explored, except under conditions of injury where, for the most part, only single components or tissue samples have been studied. Whereas maintenance or timely re-establishment of flow reduces tissue and neuron injury in both humans and animal models, protection of neuron function in humans has not prevented the evolution of injury despite the inherent mechanisms of neurovascular coupling. However, occlusion of flow to the brain rapidly identifies regions of neuron-vascular vulnerability within the vascular territory-at-risk. These coalesce to become the mature ischaemic lesion. The failure, so far, of clinical trials of neuron protectant agents to achieve detectable tissue salvage could be explained by the vulnerability (and lack of protection) of essential components of the 'unit'. This presentation summarizes evidence and thoughts on this topic. These support the need to understand component interactions within the neurovascular unit.
Collapse
Affiliation(s)
- G J del Zoppo
- Department of Medicine, University of Washington, Seattle, WA 98104, USA.
| |
Collapse
|
90
|
Miller AM, Stella N. Microglial cell migration stimulated by ATP and C5a involve distinct molecular mechanisms: quantification of migration by a novel near-infrared method. Glia 2009; 57:875-83. [PMID: 19053059 DOI: 10.1002/glia.20813] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Microglial cells, the macrophages of the brain, play an essential role in the propagation of neuroinflammation. Increased microglial cell migration in response to specific chemoattractants has been documented, but less is known about the differences between these stimuli and the signal transduction pathways that mediate their effects. Current methods to measure cell migration are often labor-intensive and rely on the manual counting of cell number, so more efficient and objective methods are needed. Here we present an improved and higher-throughput Boyden chamber technique that measures microglial cell migration by using DRAQ5, a nuclear dye that emits in the near-infrared. Out of a panel of chemoattractants tested, we found that ATP and C5a potently stimulate the migration of mouse primary microglial cells. The stimulatory effects of ATP and C5a displayed significant additivity, suggesting that each chemoattractant stimulated migration through independent molecular mechanisms. Accordingly, we found key differences in these responses: ATP stimulated a combination of both chemokinesis and chemotaxis, and this response was mediated by the ROCK signaling pathway; whereas C5a stimulated only chemotaxis and this response was mediated by the Rac1 signaling pathway. Finally, we found that functional PI3-kinase is only required for random basal microglial cell migration. Thus, our results show that distinct nonoverlapping signal transduction pathways control different modes of microglial cell migration and suggest that the targeting of these distinct molecular mechanisms should modulate different aspects of neuroinflammation propagation.
Collapse
Affiliation(s)
- Aaron M Miller
- Department of Pharmacology, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | | |
Collapse
|
91
|
Milner R. Microglial expression of alphavbeta3 and alphavbeta5 integrins is regulated by cytokines and the extracellular matrix: beta5 integrin null microglia show no defects in adhesion or MMP-9 expression on vitronectin. Glia 2009; 57:714-23. [PMID: 18985734 DOI: 10.1002/glia.20799] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
As the primary immune effector cells in the CNS, microglia play a central role in regulating inflammation. The extracellular matrix (ECM) protein vitronectin is a strong inducer of microglial activation, switching microglia from a resting into an activated potentially destructive phenotype. As the activating effect of vitronectin is mediated by alphav integrins, the aim of the current study was to evaluate the requirement of the alphavbeta5 integrin in mediating microglial adhesion and activation to vitronectin, by studying these events in beta5 integrin-null murine microglia. Surprisingly, beta5 integrin null microglia were not defective in adhesion to vitronectin. Further analysis showed that microglia express the alphavbeta3 integrin, in addition to alphavbeta5. Flow cytometry revealed that microglial alphav integrin expression is regulated by cytokines and ECM proteins. alphavbeta3 integrin expression was downregulated by IFN-gamma, TNF, LPS, and TGF-beta1. alphavbeta5 expression was also reduced by IFN-gamma, TNF, and LPS, but strongly increased by the antiactivating factors TGF-beta1 and laminin. Gel zymography revealed that beta5 integrin null microglia showed no deficiency in their expression of matrix metalloproteinase (MMP)-9 in response to vitronectin. Taken together, these data show that microglia express two different alphav integrins, alphavbeta3 and alphavbeta5, and that expression of these integrins is independently regulated by cytokines and ECM proteins. Furthermore, it reveals that the alphavbeta5 integrin is not essential for mediating microglial adhesion and MMP-9 expression in response to vitronectin.
Collapse
Affiliation(s)
- Richard Milner
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA.
| |
Collapse
|
92
|
Röhl C, Grell M, Maser E. The organotin compounds trimethyltin (TMT) and triethyltin (TET) but not tributyltin (TBT) induce activation of microglia co-cultivated with astrocytes. Toxicol In Vitro 2009; 23:1541-7. [PMID: 19422909 DOI: 10.1016/j.tiv.2009.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 04/04/2009] [Accepted: 04/28/2009] [Indexed: 10/20/2022]
Abstract
The organotin compounds trimethyltin (TMT), triethyltin (TET) and tributyltin (TBT) show different organotoxicities in vivo. While TMT and TET induce a strong neurotoxicity accompanied by microglial and astroglial activation, TBT rather effects the immune system. Previously, we have shown in an in vitro co-culture model that microglial cells can be activated by TMT in the presence of astrocytes. In this study, we wanted to investigate (a) if the neurotoxic organotin compound TET can also activate microglial cells in vitro similar to TMT and (b) if differences between the neurotoxicants TMT and TET on the one side and TBT on the other exist concerning microglial activation. Therefore, purified microglial and astroglial cell cultures from neonatal rat brains were treated either alone or in co-cultures for 24h with different concentrations of TMT, TET or TBT and the basal cytotoxicity and nitric oxide formation was determined. Furthermore, morphological changes of astrocytes were examined. Our results show that microglial activation can be increased in subcytolethal concentrations, but only in the presence of astrocytes and not in microglial cell cultures alone. This increase was induced by the neurotoxicants TMT and TET but not by TBT. Taken together, the differing microglia activating effect of the organotin compounds may contribute to the differing neurotoxic potential of this group of chemicals in vivo. In addition, our results emphasize the need for co-culture systems when studying interactions between different cell types for toxicity assessment.
Collapse
Affiliation(s)
- C Röhl
- Institute of Toxicology and Pharmacology for Natural Scientists, Christian-Albrechts-University of Kiel, Brunswiker Str. 10, D-24105 Kiel, Germany.
| | | | | |
Collapse
|
93
|
Crocker SJ, Frausto RF, Whitton JL, Milner R. A novel method to establish microglia-free astrocyte cultures: comparison of matrix metalloproteinase expression profiles in pure cultures of astrocytes and microglia. Glia 2008; 56:1187-98. [PMID: 18449943 DOI: 10.1002/glia.20689] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increased matrix metalloproteinase (MMP) proteolytic activity contributes to the pathogenesis of many neuroinflammatory and neurodegenerative conditions in the CNS. To fully understand this process, it is important to define the MMP expression profile of specific cell types, including the CNS-resident cells astrocytes and microglia. While previous studies have characterized astrocyte MMP expression by using mixed glial cultures, these results are likely complicated by the presence of contaminating microglia within these cultures. In the current study, we sought to clarify this complexity, by taking a novel approach to prepare pure astrocyte cultures entirely devoid of microglia, by promoting neural stem cell (NSC) differentiation into astrocytes. The MMP expression profile of mixed glial cultures, neurosphere-derived astrocytes, and pure microglia was characterized by RNase protection assay. This revealed that MMP gene expression is largely cell-type specific. Astrocytes constitutively expressed MMP-11, MMP-14, and MMP-2 and showed induction of MMP-3 in response to IL-1beta but did not respond to lipopolysaccharide (LPS). In contrast, microglia constitutively expressed high levels of MMP-12 and showed strong induction of MMP-9 and MMP-14 in response to LPS. Gelatin zymography confirmed that LPS and TNF-alpha induced strong expression of MMP-9 in microglia but not astrocytes. In summary, these studies demonstrate that neurosphere-derived astrocytes represent an attractive alternative system in which to study astrocyte behavior in vitro. Using this system, we have shown that astrocytes and microglia express distinct sets of MMP genes and that microglia, not astrocytes, are the major source of MMP-9 in response to LPS or TNF-alpha.
Collapse
Affiliation(s)
- Stephen J Crocker
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | |
Collapse
|
94
|
Kim H, Moon C, Kim J, Ahn M, Hyun JW, Park JW, Kim SH, Kim S, Shin T. Increased phosphorylation of caveolin-1 in the spinal cord of irradiated rats. J Vet Sci 2008; 8:323-7. [PMID: 17993744 PMCID: PMC2868146 DOI: 10.4142/jvs.2007.8.4.323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phosphorylation of caveolin-1 occurs during cell activation by various stimuli. In this study, the involvement of caveolin-1 in an irradiation injured spinal cord was examined by analyzing the phosphorylation of caveolin-1 in the spinal cord of rats after irradiation with a single dose of 15 Gray from a 60Co γ-ray source at 24 h post-irradiation (PI). A Western blot analysis showed that the phosphorylated form of caveolin-1 (p-caveolin-1) was expressed constitutively in the normal spinal cords and was significantly higher in the spinal cord of irradiated rats at 24 h PI. The increased expression of ED1, which is a marker of activated microglia/macrophages, was matched with that of p-caveolin-1. In the irradiated spinal cords, there was a higher level of p-caveolin-1 immunoreactivity in the isolectin B4-positive microglial, ependymal, and vascular endothelial cells, in which p-caveolin-1 was weakly and constitutively expressed in the normal control spinal cords. These results suggest that total body irradiation induces activation of microglial cells in the spinal cord through the phosphorylation of caveolin-1.
Collapse
Affiliation(s)
- Heechul Kim
- Department of Veterinary Medicine, College of Applied Life Sciences, Cheju National University, Jeju 690-756, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
An α5β1 integrin inhibitor attenuates glioma growth. Mol Cell Neurosci 2008; 39:579-85. [DOI: 10.1016/j.mcn.2008.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 07/24/2008] [Accepted: 08/13/2008] [Indexed: 11/19/2022] Open
|
96
|
Wakselman S, Béchade C, Roumier A, Bernard D, Triller A, Bessis A. Developmental neuronal death in hippocampus requires the microglial CD11b integrin and DAP12 immunoreceptor. J Neurosci 2008; 28:8138-43. [PMID: 18685038 PMCID: PMC6670768 DOI: 10.1523/jneurosci.1006-08.2008] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2008] [Revised: 05/29/2008] [Accepted: 06/28/2008] [Indexed: 12/28/2022] Open
Abstract
In several brain regions, microglia actively promote neuronal apoptosis during development. However, molecular actors leading microglia to trigger death remain mostly unknown. Here, we show that, in the developing hippocampus, apoptotic neurons are contacted by microglia expressing both the integrin CD11b and the immunoreceptor DAP12. We demonstrate that developmental apoptosis decreases in mice deficient for CD11b or DAP12. In addition, function-blocking antibodies directed against CD11b decrease neuronal death when injected into wild-type neonates, but have no effect when injected into DAP12-deficient littermates. This demonstrates that DAP12 and CD11b act in converging pathways to induce neuronal death. Finally, we show that DAP12 and CD11b control the production of microglial superoxide ions, which kill the neurons. Thus, our data show that the process of developmental neuronal death triggered by microglia is similar to the elimination of pathogenic cells by the innate immune cells.
Collapse
Affiliation(s)
- Shirley Wakselman
- Laboratoire de Biologie Cellulaire de la Synapse, Institut National de la Santé et de la Recherche Médicale, Unité 789, 75230 Paris Cedex 05, France
| | - Catherine Béchade
- Laboratoire de Biologie Cellulaire de la Synapse, Institut National de la Santé et de la Recherche Médicale, Unité 789, 75230 Paris Cedex 05, France
| | - Anne Roumier
- Laboratoire de Biologie Cellulaire de la Synapse, Institut National de la Santé et de la Recherche Médicale, Unité 789, 75230 Paris Cedex 05, France
| | - Delphine Bernard
- Laboratoire de Biologie Cellulaire de la Synapse, Institut National de la Santé et de la Recherche Médicale, Unité 789, 75230 Paris Cedex 05, France
| | - Antoine Triller
- Laboratoire de Biologie Cellulaire de la Synapse, Institut National de la Santé et de la Recherche Médicale, Unité 789, 75230 Paris Cedex 05, France
| | - Alain Bessis
- Laboratoire de Biologie Cellulaire de la Synapse, Institut National de la Santé et de la Recherche Médicale, Unité 789, 75230 Paris Cedex 05, France
| |
Collapse
|
97
|
Tsuda M, Toyomitsu E, Komatsu T, Masuda T, Kunifusa E, Nasu-Tada K, Koizumi S, Yamamoto K, Ando J, Inoue K. Fibronectin/integrin system is involved in P2X(4) receptor upregulation in the spinal cord and neuropathic pain after nerve injury. Glia 2008; 56:579-85. [PMID: 18240312 DOI: 10.1002/glia.20641] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We have previously shown that activation of the ATP-gated ion channel subtype P2X(4) receptors (P2X(4)Rs) in the spinal cord, the expression of which is upregulated in microglia after nerve injury, is necessary for producing neuropathic pain. The upregulation of P2X(4)Rs in microglia is, therefore, a key process in neuropathic pain, but the mechanism remains unknown. Here, we find a fibronectin/integrin-dependent mechanism in the upregulation of P2X(4)Rs. Microglia cultured on dishes coated with fibronectin, an extracellular matrix molecule, expressed a higher level of P2X(4)R protein when compared with those cultured on control dishes. The increase was suppressed by echistatin, a peptide that selectively blocks beta(1) and beta(3)-containing integrins, and with a function-blocking antibody of beta(1) integrin. In in vivo studies, the upregulation of P2X(4)Rs in the spinal cord after spinal nerve injury was significantly suppressed by intrathecal administration of echistatin. Tactile allodynia in response to nerve injury and intrathecal administration of ATP- and fibronectin-stimulated microglia was inhibited by echistatin. Furthermore, intrathecal administration of fibronectin in normal rats increased the level of P2X(4)R protein in the spinal cord and produced tactile allodynia. Moreover, the fibronectin-induced allodynia was not observed in mice lacking P2X(4)R. Taken together with the results of our previous study showing an increase in the spinal fibronectin level after nerve injury, the present results suggest that the fibronectin/integrin system participates in the upregulation of P2X(4)R expression after nerve injury and subsequent neuropathic pain.
Collapse
Affiliation(s)
- Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Hermann PM, Nicol JJ, Bulloch AGM, Wildering WC. RGD-dependent mechanisms in the endoneurial phagocyte response and axonal regeneration in the nervous system of the snail Lymnaea stagnalis. ACTA ACUST UNITED AC 2008; 211:491-501. [PMID: 18245625 DOI: 10.1242/jeb.013102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of phagocytic cells in the injury zone is a crucial step in the regeneration of peripheral axons. Many aspects of the mechanisms underlying the recruitment of active phagocytes remain, however, unclear. Notably, our understanding of the interactions between injury, extracellular matrix (ECM) degradation and phagocyte activation is limited. Most animal cell types, phagocytes included, interact with proteins of the ECM through one or more members of the integrin family, transmembrane cell adhesion receptors that typically bind their ligands through short linear amino acid sequences. This study focused on the role of one of the most common of such integrin recognition sequences, the Arg-Gly-Asp (RGD) motif in the recruitment and activation of endoneurial phagocytes in the injury response of the nervous system of the pond snail Lymnaea stagnalis. Like the mammalian nervous system, the Lymnaea nervous system responds to injury with recruitment and activation of endoneurial phagocytes (i.e. phagocytes residing in Lymnaea's nerves), a process involving substantial changes in the morphology, motility and adhesion status of these cells. Using synthetic water-soluble RGD-peptides, we investigated the relevance of RGD-dependent mechanisms in the activation of endoneurial phagocytes and injury response of the organ-cultured nervous system of Lymnaea. Our results show that RGD-peptides modulate various aspects of phagocyte activation (i.e. spreading response, particle engulfment, oxidative burst) in vitro and in situ and significantly affect nerve regeneration in this model system. Surprisingly, while linear RGD-analogues suppressed both phagocyte activation and axonal regeneration, a circularized RGD-peptide analogue modulated these parameters in a concentration-dependent, biphasic manner. Collectively, these results emphasize the significance of RGD-dependent mechanisms in the regenerative response of the Lymnaea nervous system and implicate regulation of the cellular immune response as one of the factors in this context.
Collapse
Affiliation(s)
- Petra M Hermann
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada, T2N 1N4
| | | | | | | |
Collapse
|
99
|
Coyne TM, Marcus AJ, Reynolds K, Black IB, Woodbury D. Disparate host response and donor survival after the transplantation of mesenchymal or neuroectodermal cells to the intact rodent brain. Transplantation 2008; 84:1507-16. [PMID: 18091528 DOI: 10.1097/01.tp.0000288185.09601.4d] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND To circumvent ethical and legal complications associated with embryonic cell sources, investigators have proposed the use of nonneural donor sources for use in neural transplantation strategies. Leading candidate sources include autologous marrow stromal cells (MSCs) and fibroblasts, which are mesodermal derivatives. However, we recently reported that MSCs transplanted to the adult brain are rapidly rejected by an inflammatory response. Whether extrinsic variables or intrinsic mesenchymal traits stimulated inflammation and rejection is unknown. To determine the future utility of these cells in neural transplantation, we have now performed a systematic analysis of MSC transplantation to the brain. METHODS To examine the effects of extrinsic variables on transplantation, green fluorescent protein (GFP)-expressing rat MSCs, cultured under distinct conditions, were transplanted stereotactically to the normal adult rat striatum, and donor survival and the host response was compared. To examine whether intrinsic donor traits promoted rejection, 50,000 GFP-expressing rat MSCs, fibroblasts, or astrocytes were transplanted stereotactically to the adult rat striatum and graft survival and the host response was compared. RESULTS Irrespective of preoperative culture conditions, MSCs elicited an inflammatory response and were rejected by 14 days, indicating acute rejection was not mediated by culture conditions. Comparison of MSC, fibroblast, or astrocyte grafts revealed that mesenchymal derivatives, MSCs and fibroblasts, elicited an inflammatory response and were rapidly rejected, whereas neuroectodermal astrocytes demonstrated robust survival in the absence of inflammation. CONCLUSIONS Our findings suggest that intrinsic characteristics of mesenchymal cells may stimulate host inflammation, and thus may not represent an ideal donor source for transplantation to the adult brain.
Collapse
Affiliation(s)
- Thomas M Coyne
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| | | | | | | | | |
Collapse
|
100
|
Cytokines and Extracellular Matrix Remodeling in the Central Nervous System. CYTOKINES AND THE BRAIN 2008. [DOI: 10.1016/s1567-7443(07)10009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|