Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Neuroinflammation ; 13(1): 249, 2016 Sep 21.
Article in English | MEDLINE | ID: mdl-27654972

ABSTRACT

BACKGROUND: Neuroinflammation and blood-brain barrier (BBB) disruption are common features of many brain disorders, including Alzheimer's disease, epilepsy, and motor neuron disease. Inflammation is thought to be a driver of BBB breakdown, but the underlying mechanisms for this are unclear. Brain pericytes are critical cells for maintaining the BBB and are immunologically active. We sought to test the hypothesis that inflammation regulates the BBB by altering pericyte biology. METHODS: We exposed primary adult human brain pericytes to chronic interferon-gamma (IFNγ) for 4 days and measured associated functional aspects of pericyte biology. Specifically, we examined the influence of inflammation on platelet-derived growth factor receptor-beta (PDGFRß) expression and signalling, as well as pericyte proliferation and migration by qRT-PCR, immunocytochemistry, flow cytometry, and western blotting. RESULTS: Chronic IFNγ treatment had marked effects on pericyte biology most notably through the PDGFRß, by enhancing agonist (PDGF-BB)-induced receptor phosphorylation, internalization, and subsequent degradation. Functionally, chronic IFNγ prevented PDGF-BB-mediated pericyte proliferation and migration. CONCLUSIONS: Because PDGFRß is critical for pericyte function and its removal leads to BBB leakage, our results pinpoint a mechanism linking chronic brain inflammation to BBB dysfunction.

2.
J Neuroinflammation ; 13: 37, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26867675

ABSTRACT

BACKGROUND: Transforming growth factor beta 1 (TGFß1) is strongly induced following brain injury and polarises microglia to an anti-inflammatory phenotype. Augmentation of TGFß1 responses may therefore be beneficial in preventing inflammation in neurological disorders including stroke and neurodegenerative diseases. However, several other cell types display immunogenic potential and identifying the effect of TGFß1 on these cells is required to more fully understand its effects on brain inflammation. Pericytes are multifunctional cells which ensheath the brain vasculature and have garnered recent attention with respect to their immunomodulatory potential. Here, we sought to investigate the inflammatory phenotype adopted by TGFß1-stimulated human brain pericytes. METHODS: Microarray analysis was performed to examine transcriptome-wide changes in TGFß1-stimulated pericytes, and results were validated by qRT-PCR and cytometric bead arrays. Flow cytometry, immunocytochemistry and LDH/Alamar Blue® viability assays were utilised to examine phagocytic capacity of human brain pericytes, transcription factor modulation and pericyte health. RESULTS: TGFß1 treatment of primary human brain pericytes induced the expression of several inflammatory-related genes (NOX4, COX2, IL6 and MMP2) and attenuated others (IL8, CX3CL1, MCP1 and VCAM1). A synergistic induction of IL-6 was seen with IL-1ß/TGFß1 treatment whilst TGFß1 attenuated the IL-1ß-induced expression of CX3CL1, MCP-1 and sVCAM-1. TGFß1 was found to signal through SMAD2/3 transcription factors but did not modify nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) translocation. Furthermore, TGFß1 attenuated the phagocytic ability of pericytes, possibly through downregulation of the scavenger receptors CD36, CD47 and CD68. Whilst TGFß did decrease pericyte number, this was due to a reduction in proliferation, not apoptotic death or compromised cell viability. CONCLUSIONS: TGFß1 attenuated pericyte expression of key chemokines and adhesion molecules involved in CNS leukocyte trafficking and the modulation of microglial function, as well as reduced the phagocytic ability of pericytes. However, TGFß1 also enhanced the expression of classical pro-inflammatory cytokines and enzymes which can disrupt BBB functioning, suggesting that pericytes adopt a phenotype which is neither solely pro- nor anti-inflammatory. Whilst the effects of pericyte modulation by TGFß1 in vivo are difficult to infer, the reduction in pericyte proliferation together with the elevated IL-6, MMP-2 and NOX4 and reduced phagocytosis suggests a detrimental action of TGFß1 on neurovasculature.


Subject(s)
Brain/cytology , Cytokines/metabolism , Gene Expression Regulation/drug effects , Pericytes/drug effects , Phagocytes/drug effects , Transforming Growth Factor beta1/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/metabolism , Humans , Interleukin-1beta/pharmacology , Matrix Metalloproteinase 2/metabolism , NADPH Oxidase 4 , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Time Factors , Vascular Cell Adhesion Molecule-1/metabolism
3.
J Neuroinflammation ; 11: 104, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24920309

ABSTRACT

BACKGROUND: Brain inflammation plays a key role in neurological disease. Although much research has been conducted investigating inflammatory events in animal models, potential differences in human brain versus rodent models makes it imperative that we also study these phenomena in human cells and tissue. METHODS: Primary human brain cell cultures were generated from biopsy tissue of patients undergoing surgery for drug-resistant epilepsy. Cells were treated with pro-inflammatory compounds IFNγ, TNFα, IL-1ß, and LPS, and chemokines IP-10 and MCP-1 were measured by immunocytochemistry, western blot, and qRT-PCR. Microarray analysis was also performed on late passage cultures treated with vehicle or IFNγ and IL-1ß. RESULTS: Early passage human brain cell cultures were a mixture of microglia, astrocytes, fibroblasts and pericytes. Later passage cultures contained proliferating fibroblasts and pericytes only. Under basal culture conditions all cell types showed cytoplasmic NFκB indicating that they were in a non-activated state. Expression of IP-10 and MCP-1 were significantly increased in response to pro-inflammatory stimuli. The two chemokines were expressed in mixed cultures as well as cultures of fibroblasts and pericytes only. The expression of IP-10 and MCP-1 were regulated at the mRNA and protein level, and both were secreted into cell culture media. NFκB nuclear translocation was also detected in response to pro-inflammatory cues (except IFNγ) in all cell types. Microarray analysis of brain pericytes also revealed widespread changes in gene expression in response to the combination of IFNγ and IL-1ß treatment including interleukins, chemokines, cellular adhesion molecules and much more. CONCLUSIONS: Adult human brain cells are sensitive to cytokine challenge. As expected 'classical' brain immune cells, such as microglia and astrocytes, responded to cytokine challenge but of even more interest, brain pericytes also responded to such challenge with a rich repertoire of gene expression. Immune activation of brain pericytes may play an important role in communicating inflammatory signals to and within the brain interior and may also be involved in blood brain barrier (BBB) disruption . Targeting brain pericytes, as well as microglia and astrocytes, may provide novel opportunities for reducing brain inflammation and maintaining BBB function and brain homeostasis in human brain disease.


Subject(s)
Brain/pathology , Cytokines/metabolism , Cytokines/pharmacology , Pericytes/drug effects , Pericytes/metabolism , Actins/metabolism , Adult , Antigens/metabolism , Cells, Cultured , Cytokines/genetics , Dura Mater/drug effects , Dura Mater/metabolism , Epilepsy/pathology , Fibronectins/metabolism , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Leukocyte Common Antigens/metabolism , Male , Middle Aged , Neuroglia/drug effects , Organ Culture Techniques , Protein Transport/drug effects , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Time Factors , Transcription Factor RelA/metabolism
4.
Glia ; 61(6): 929-42, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23483680

ABSTRACT

Microglia are the predominant resident immune cells of the brain and can assume a range of phenotypes. They are critical for normal brain development and function but can also contribute to many disease processes. Although they are widely studied, the transcriptional control of microglial phenotype and activation requires further research. PU.1 is a key myeloid transcription factor expressed by peripheral macrophages and rodent microglia. In this article, we report the presence of PU.1 specifically in microglia of the adult human brain and we examine its functional role in primary human microglia. Using siRNA, we achieved substantial PU.1 protein knock-down in vitro. By assessing a range of characteristic microglial proteins we found decreased viability of adult human microglia with reduced PU.1 protein expression. This observation was confirmed with PU.1 antisense DNA oligonucleotides. An important function of microglia is to clear debris by phagocytosis. We assessed the impact of loss of PU.1 on microglial phagocytosis and show that PU.1 siRNA reduces the ability of adult human microglia to phagocytose amyloid-beta1-42 peptide. These results show that PU.1 controls human microglial viability and function and suggest PU.1 as a molecular target for manipulation of human microglial phenotype.


Subject(s)
Brain/metabolism , Cell Survival/physiology , Microglia/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Phagocytosis/physiology , Proto-Oncogene Proteins/genetics , RNA, Small Interfering , Trans-Activators/genetics
5.
J Neuroinflammation ; 10: 85, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23866312

ABSTRACT

BACKGROUND: Microglia are the primary immune cells of the brain whose phenotype largely depends on their surrounding micro-environment. Microglia respond to a multitude of soluble molecules produced by a variety of brain cells. Macrophage colony-stimulating factor (M-CSF) is a cytokine found in the brain whose receptor is expressed by microglia. Previous studies suggest a critical role for M-CSF in brain development and normal functioning as well as in several disease processes involving neuroinflammation. METHODS: Using biopsy tissue from patients with intractable temporal epilepsy and autopsy tissue, we cultured primary adult human microglia to investigate their response to M-CSF. Mixed glial cultures were treated with 25 ng/ml M-CSF for 96 hours. Proliferation and phagocytosis assays, and high through-put immunocytochemistry, microscopy and image analysis were performed to investigate microglial phenotype and function. RESULTS: We found that the phenotype of primary adult human microglia was markedly changed following exposure to M-CSF. A greater number of microglia were present in the M-CSF- treated cultures as the percentage of proliferating (BrdU and Ki67-positive) microglia was greatly increased. A number of changes in protein expression occurred following M-CSF treatment, including increased transcription factors PU.1 and C/EBPß, increased DAP12 adaptor protein, increased M-CSF receptor (CSF-1R) and IGF-1 receptor, and reduced HLA-DP, DQ, DR antigen presentation protein. Furthermore, a distinct morphological change was observed with elongation of microglial processes. These changes in phenotype were accompanied by a functional increase in phagocytosis of Aß1-42 peptide. CONCLUSIONS: We show here that the cytokine M-CSF dramatically influences the phenotype of adult human microglia. These results pave the way for future investigation of M-CSF-related targets for human therapeutic benefit.


Subject(s)
Cell Proliferation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Microglia/drug effects , Phagocytosis/drug effects , Transcription Factors/biosynthesis , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Antimetabolites , Autopsy , Biopsy , Bromodeoxyuridine , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Cells, Cultured , HLA Antigens/biosynthesis , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Ki-67 Antigen/metabolism , Macrophage Activation/drug effects , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microglia/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Trans-Activators/biosynthesis , Trans-Activators/genetics
6.
Front Cell Neurosci ; 16: 1047928, 2022.
Article in English | MEDLINE | ID: mdl-36425665

ABSTRACT

The study of microglia isolated from adult human brain tissue provides unique insight into the physiology of these brain immune cells and their role in adult human brain disorders. Reports of microglia in post-mortem adult human brain tissue show regional differences in microglial populations, however, these differences have not been fully explored in living microglia. In this study biopsy tissue was obtained from epileptic patients undergoing surgery and consisted of both cortical areas and neurogenic ventricular and hippocampal (Hp) areas. Microglia were concurrently isolated from both regions and compared by immunochemistry. Our initial observation was that a greater number of microglia resulted from isolation and culture of ventricular/Hp tissue than cortical tissue. This was found to be due to a greater proliferative capacity of microglia from ventricular/Hp regions compared to the cortex. Additionally, ventricular/Hp microglia had a greater proliferative response to the microglial mitogen Macrophage Colony-Stimulating Factor (M-CSF). This enhanced response was found to be associated with higher M-CSF receptor expression and higher expression of proteins involved in M-CSF signalling DAP12 and C/EBPß. Microglia from the ventricular/Hp region also displayed higher expression of the receptor for Insulin-like Growth Factor-1, a molecule with some functional similarity to M-CSF. Compared to microglia isolated from the cortex, ventricular/Hp microglia showed increased HLA-DP, DQ, DR antigen presentation protein expression and a rounded morphology. These findings show that microglia from adult human brain neurogenic regions are more proliferative than cortical microglia and have a distinct protein expression profile. The data present a case for differential microglial phenotype and function in different regions of the adult human brain and suggest that microglia in adult neurogenic regions are "primed" to an activated state by their unique tissue environment.

7.
Neurooncol Adv ; 3(1): vdab031, 2021.
Article in English | MEDLINE | ID: mdl-34286275

ABSTRACT

BACKGROUND: Microglia and tumor-associated macrophages (TAMs) constitute up to half of the total tumor mass of glioblastomas. Despite these myeloid populations being ontogenetically distinct, they have been largely conflated. Recent single-cell transcriptomic studies have identified genes that distinguish microglia from TAMs. Here we investigated whether the translated proteins of genes enriched in microglial or TAM populations can be used to differentiate these myeloid cells in immunohistochemically stained human glioblastoma tissue. METHODS: Tissue sections from resected low-grade, meningioma, and glioblastoma (grade IV) tumors and epilepsy tissues were immunofluorescently triple-labeled for Iba1 (pan-myeloid marker), CD14 or CD163 (preferential TAM markers), and either P2RY12 or TMEM119 (microglial-specific markers). Using a single-cell-based image analysis pipeline, we quantified the abundance of each marker within single myeloid cells, allowing the identification and analysis of myeloid populations. RESULTS: P2RY12 and TMEM119 successfully discriminated microglia from TAMs in glioblastoma. In contrast, CD14 and CD163 expression were not restricted to invading TAMs and were upregulated by tumor microglia. Notably, a higher ratio of microglia to TAMs significantly correlated with increased patient survival. CONCLUSIONS: We demonstrate the validity of previously defined microglial-specific genes P2RY12 and TMEM119 as robust discriminators of microglia and TAMs at the protein level in human tissue. Moreover, our data suggest that a higher proportion of microglia may be beneficial for patient survival in glioblastoma. Accordingly, this tissue-based method for myeloid population differentiation could serve as a useful prognostic tool.

9.
Sci Rep ; 6: 19371, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26778406

ABSTRACT

Microglia, the resident macrophages of the central nervous system play vital roles in brain homeostasis through clearance of pathogenic material. Microglia are also implicated in neurological disorders through uncontrolled activation and inflammatory responses. To date, the vast majority of microglial studies have been performed using rodent models. Human microglia differ from rodent counterparts in several aspects including their response to pharmacological substances and their inflammatory secretions. Such differences highlight the need for studies on primary adult human brain microglia and methods to isolate them are therefore required. Our procedure generates microglial cultures of >95% purity from both biopsy and autopsy human brain tissue using a very simple media-based culture procedure that takes advantage of the adherent properties of these cells. Microglia obtained in this manner can be utilised for research within a week. Isolated microglia demonstrate phagocytic ability and respond to inflammatory stimuli and their purity makes them suitable for numerous other forms of in vitro studies, including secretome and transcriptome analysis. Furthermore, this protocol allows for the simultaneous isolation of neural precursor cells during the microglial isolation procedure. As human brain tissue is such a precious and valuable resource the simultaneous isolation of multiple cell types is highly beneficial.


Subject(s)
Cell Separation , Microglia/cytology , Microglia/physiology , Biomarkers , Cell Separation/methods , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Gene Expression Regulation , Humans , Immunohistochemistry , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Phagocytosis , Phenotype , Protein Transport
10.
Sci Rep ; 5: 12132, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26166618

ABSTRACT

Neuroinflammation contributes to the pathogenesis of several neurological disorders and pericytes are implicated in brain inflammatory processes. Cellular inflammatory responses are orchestrated by transcription factors but information on transcriptional control in pericytes is lacking. Because the transcription factor CCAAT/enhancer binding protein delta (C/EBPδ) is induced in a number of inflammatory brain disorders, we sought to investigate its role in regulating pericyte immune responses. Our results reveal that C/EBPδ is induced in a concentration- and time-dependent fashion in human brain pericytes by interleukin-1ß (IL-1ß). To investigate the function of the induced C/EBPδ in pericytes we used siRNA to knockdown IL-1ß-induced C/EBPδ expression. C/EBPδ knockdown enhanced IL-1ß-induced production of intracellular adhesion molecule-1 (ICAM-1), interleukin-8, monocyte chemoattractant protein-1 (MCP-1) and IL-1ß, whilst attenuating cyclooxygenase-2 and superoxide dismutase-2 gene expression. Altered ICAM-1 and MCP-1 protein expression were confirmed by cytometric bead array and immunocytochemistry. Our results show that knock-down of C/EBPδ expression in pericytes following immune stimulation increased chemokine and adhesion molecule expression, thus modifying the human brain pericyte inflammatory response. The induction of C/EBPδ following immune stimulation may act to limit infiltration of peripheral immune cells, thereby preventing further inflammatory responses in the brain.


Subject(s)
Anti-Inflammatory Agents/metabolism , CCAAT-Enhancer-Binding Protein-delta/metabolism , Inflammation/metabolism , Pericytes/metabolism , Brain/metabolism , Brain/pathology , Gene Expression Regulation/physiology , Humans , Inflammation/pathology , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Pericytes/pathology , Superoxide Dismutase/metabolism , Transcription Factors/metabolism
11.
PLoS One ; 8(12): e80463, 2013.
Article in English | MEDLINE | ID: mdl-24339874

ABSTRACT

The chemokine Interferon gamma-induced protein 10 (IP-10) and human leukocyte antigen (HLA) are widely used indicators of glial activation and neuroinflammation and are up-regulated in many brain disorders. These inflammatory mediators have been widely studied in rodent models of brain disorders, but less work has been undertaken using human brain cells. In this study we investigate the regulation of HLA and IP-10, as well as other cytokines and chemokines, in microglia, astrocytes, pericytes, and meningeal fibroblasts derived from biopsy and autopsy adult human brain, using immunocytochemistry and a Cytometric Bead Array. Interferonγ (IFNγ) increased microglial HLA expression, but contrary to data in rodents, the anti-inflammatory cytokine transforming growth factor ß1 (TGFß1) did not inhibit this increase in HLA, nor did TGFß1 affect basal microglial HLA expression or IFNγ-induced astrocytic HLA expression. In contrast, IFNγ-induced and basal microglial HLA expression, but not IFNγ-induced astrocytic HLA expression, were strongly inhibited by macrophage colony stimulating factor (M-CSF). IFNγ also strongly induced HLA expression in pericytes and meningeal fibroblasts, which do not basally express HLA, and this induction was completely blocked by TGFß1, but not affected by M-CSF. In contrast, TGFß1 did not block the IFNγ-induced increase in IP-10 in pericytes and meningeal fibroblasts. These results show that IFNγ, TGFß1 and M-CSF have species- and cell type-specific effects on human brain cells that may have implications for their roles in adult human brain inflammation.


Subject(s)
Fibroblasts/drug effects , Interferon-gamma/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Meninges/cytology , Neuroglia/cytology , Pericytes/cytology , Transforming Growth Factor beta1/pharmacology , Adult , Astrocytes/cytology , Chemokines/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HLA-D Antigens/metabolism , Humans
12.
Heart Lung Circ ; 11(3): 189-92, 2002.
Article in English | MEDLINE | ID: mdl-16352096

ABSTRACT

The diagnosis of sarcoidosis of the heart can be elusive, and is seldom established in life. We report the case of a 43-year-old man who underwent heart transplantation for presumed idiopathic dilated cardiomyopathy. Endomyocardial biopsy before transplantation showed only a mild infiltrate of lymphocytes. Histology of his explanted heart revealed extensive noncaseating granulomas and scarring, typical of sarcoidosis. A diagnosis of sarcoidosis had been made several years before by mediastinoscopic biopsy, after routine chest X-ray revealed mediastinal lymphadenopathy. Aside from the cardiac manifestations, the patient had no other symptoms of this disease. We discuss the inherent difficulties in the diagnosis of this rare but important condition, its varying presentations relating to the underlying pathology, as well as treatment options, including the role of transplantation.

SELECTION OF CITATIONS
SEARCH DETAIL