Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Int J Mol Sci ; 24(7)2023 Mar 23.
Article in English | MEDLINE | ID: mdl-37047054

ABSTRACT

It is known that many cells produce extracellular vesicles, and this includes a range of different cancer cell types. Here we demonstrate the profound effects of large vesicular-like bodies produced by melanoma cells on the barrier integrity of human brain endothelial cells. These vesicular-bodies have not been fully characterised but range in size from ~500 nm to >10 µm, are surrounded by membrane and are enzymatically active based on cell-tracker incorporation. Their size is consistent with previously reported large oncosomes and apoptotic bodies. We demonstrate that these melanoma-derived vesicular-bodies rapidly affect brain endothelial barrier integrity, measured using ECIS biosensor technology, where the disruption is evident within ~60 min. This disruption involves acquisition of the vesicles through transcellular uptake into the endothelial cells. We also observed extensive actin-rearrangement, actin removal from the paracellular boundary of the endothelial cells and envelopment of the vesicular-bodies by actin. This was concordant with widespread changes in CD144 localisation, which was consistent with the loss of junctional strength. High-resolution confocal imaging revealed proximity of the melanoma vesicular-bodies juxtaposed to the endothelial nucleus, often containing fragmented DNA themselves, raising speculation over this association and potential delivery of nuclear material into the brain endothelial cells. The disruption of the endothelial cells occurs in a manner that is faster and completely distinct to that of invasion by intact melanoma cells. Given the clinical observation of large vesicles in the circulation of melanoma patients by others, we hypothesize their involvement in weakening or priming the brain vasculature for melanoma invasion.


Subject(s)
Endothelial Cells , Melanoma , Humans , Endothelial Cells/metabolism , Blood-Brain Barrier/metabolism , Actins/metabolism , Brain/metabolism , Melanoma/metabolism
2.
BMC Neurol ; 22(1): 216, 2022 Jun 11.
Article in English | MEDLINE | ID: mdl-35690735

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable and rapidly progressive neurological disorder. Biomarkers are critical to understanding disease causation, monitoring disease progression and assessing the efficacy of treatments. However, robust peripheral biomarkers are yet to be identified. Neuroinflammation and breakdown of the blood-brain barrier (BBB) are common to familial and sporadic ALS and may produce a unique biomarker signature in peripheral blood. Using cytometric bead array (n = 15 participants per group (ALS or control)) and proteome profiling (n = 6 participants per group (ALS or control)), we assessed a total of 106 serum cytokines, growth factors, and BBB breakdown markers in the serum of control and ALS participants. Further, primary human brain pericytes, which maintain the BBB, were used as a biosensor of inflammation following pre-treatment with ALS serum. Principal components analysis of all proteome profile data showed no clustering of control or ALS sera, and no individual serum proteins met the threshold for statistical difference between ALS and controls (adjusted P values). However, the 20 most changed proteins between control and ALS sera showed a medium effect size (Cohen's d = 0.67) and cluster analysis of their levels together identified three sample subsets; control-only, mixed control-ALS, and ALS-only. These 20 proteins were predominantly pro-angiogenic and growth factors, including fractalkine, BDNF, EGF, PDGF, Dkk-1, MIF and angiopoietin-2. S100ß, a protein highly concentrated in glial cells and therefore a marker of BBB leakage when found in blood, was unchanged in ALS serum, suggesting that serum protein profiles were reflective of peripheral rather than CNS biofluids. Finally, primary human brain pericytes remained proliferative and their secretome was unchanged by chronic exposure to ALS serum. Our exploratory study suggests that individual serum cytokine levels may not be robust biomarkers in small studies of ALS, but that larger studies using multiplexed analysis of pro-angiogenic and growth factors may identify a peripheral signature of ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/pathology , Biomarkers , Blood-Brain Barrier/metabolism , Cytokines , Humans , Intercellular Signaling Peptides and Proteins , Neuroinflammatory Diseases , Proteome/metabolism
3.
Int J Mol Sci ; 23(22)2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36430641

ABSTRACT

Glioblastoma is refractory to therapy and presents a significant oncological challenge. Promising immunotherapies have not shown the promise observed in other aggressive cancers. The reasons for this include the highly immuno-suppressive tumour microenvironment controlled by the glioblastoma cells and heterogeneous phenotype of the glioblastoma cells. Here, we wanted to better understand which glioblastoma phenotypes produced the regulatory cytokines, particularly those that are implicated in shaping the immune microenvironment. In this study, we employed nanoString analysis of the glioblastoma transcriptome, and proteomic analysis (proteome profiler arrays and cytokine profiling) of secreted cytokines by different glioblastoma phenotypes. These phenotypes were cultured to reflect a spectrum of glioblastoma cells present in tumours, by culturing an enhanced stem-like phenotype of glioblastoma cells or a more differentiated phenotype following culture with serum. Extensive secretome profiling reveals that there is considerable heterogeneity in secretion patterns between serum-derived and glioblastoma stem-like cells, as well as between individuals. Generally, however, the serum-derived phenotypes appear to be the primary producers of cytokines associated with immune cell recruitment into the tumour microenvironment. Therefore, these glioblastoma cells have considerable importance in shaping the immune landscape in glioblastoma and represent a valuable therapeutic target that should not be ignored.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/pathology , Cytokines/genetics , Brain Neoplasms/pathology , Proteomics , Phenotype , Tumor Microenvironment
4.
Cell Biol Int ; 44(1): 343-351, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31498530

ABSTRACT

Neuroinflammatory disorders such as Alzheimer's and Parkinson's diseases are characterised by chronic inflammation and loss of vascular integrity. Bradykinin 1 receptor (B1R) activation has been implicated in many neuroinflammatory diseases, but the contribution of B1R to inflammation and vascular breakdown is yet to be determined. As a result, the present study evaluated the effect of B1R stimulation using Des-Arg-9-BK on the cytokine profile and junctional properties of human cerebral microvascular endothelial cells (hCMVECs). Results showed that stimulation of B1R receptors increased secretion of pro-inflammatory cytokines, interleukin-6 (IL-6), IL-8, intracellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1), but decreased the expression of vascular endothelial growth factor (VEGF), a cytokine and growth factor required for maintenance of the vasculature. B1R stimulation also resulted in the loss of occludin expression at tight junctions with no change in VE-cadherin expression. There was also a significant increase in permeability to Evans blue albumin, suggesting an increase of vascular permeability. Taken together, these results suggest that B1R activation that occurs in neuroinflammatory diseases may contribute to both the inflammation and loss of blood-brain barrier integrity that is characteristic of these diseases.

5.
Int J Mol Sci ; 21(21)2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33139674

ABSTRACT

We have recently demonstrated that invasive melanoma cells are capable of disrupting the brain endothelial barrier integrity. This was shown using ECIS biosensor technology, which revealed rapid disruption via the paracellular junctions. In this paper, we demonstrate that melanoma cells secrete factors (e.g., cytokines) that weaken the endothelial barrier integrity. Through proteome profiling, we attempt to identify the barrier-disrupting cytokines. Melanoma conditioned media were collected from three New Zealand melanoma lines. ECIS technology was used to assess if the conditioned media disrupted the endothelial barrier independent of the melanoma cells. The melanoma cell secretome was assessed using cytometric bead array (CBA), Luminex immunoassay and multiplex Proteome Profilers, to detect the expression of secretory proteins, which may facilitate metastasis. Finally, ECIS technology was used to assess the direct effects of secreted proteins identified as candidates from the proteome screens. We show that melanoma-conditioned media significantly disrupted the brain endothelial barrier, however, to a much lesser extent than the cells from which they were collected. Cytokine and proteome profiling of the conditioned media showed evidence of high concentrations of approximately 15 secreted proteins (including osteopontin, IL-8, GDF-15, MIF and VEGF). These 15 secreted proteins were expressed variably across the melanoma lines. Surprisingly, the addition of these individually to the brain endothelial cells did not substantially affect the barrier integrity. ANGPTL-4 and TGFß were also produced by the melanoma cells. Whilst TGFß-1 had a pronounced effect on the barrier integrity, surprisingly ANGPTL-4 did not. However, its C-terminal fragment did and within a very similar period to the conditioned media, albeit not to the same extent. Herein we show that melanoma cells produce a wide-range of soluble factors at high concentrations, which most likely favour support or survival of the cancer cells. Most of these, except for TGFß-1 and the C-terminal fragment of ANGPTL-4, did not have an impact on the integrity of the brain endothelial cells.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Biosensing Techniques/methods , Blood-Brain Barrier/drug effects , Brain/pathology , Cell Line , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Flow Cytometry/methods , Humans , Immunoassay/methods , Melanoma/genetics , Melanoma/pathology , Proteome/metabolism , Proteomics/methods , Skin Neoplasms/genetics , Skin Neoplasms/pathology
6.
Biochim Biophys Acta Gen Subj ; 1862(3): 385-393, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29158134

ABSTRACT

BACKGROUND: Connexin43 hemichannels have been implicated in many inflammatory diseases including diabetic retinopathy (DR). Particularly, hemichannel-mediated ATP release has been associated with inflammasome pathway activation. Using an in vitro cell culture model, we evaluated hemichannel roles in response to inflammatory cytokines under high glucose (HG) conditions and propose a mechanism by which a connexin43 hemichannel-mediated autocrine ATP feedback loop augments chronic inflammatory disease. METHODS: Retinal pigment epithelial cells were exposed to HG, 10ng/mL pro-inflammatory cytokines IL-1ß and TNF-α, or a combination of both. Quantitative Cytometric Bead Array analysis was used to measure the release of inflammatory cytokines IL-6, IL-8, MCP-1, and sICAM-1, as well as VEGF and ATP. To determine the role of connexin43 hemichannels in the disease process, changes in cytokine and ATP release were evaluated following treatment with Peptide5, a connexin43 hemichannel blocker. Immunohistochemistry was used to compare NLRP3 inflammasome assembly under control and treatment conditions. RESULTS: Co-application of HG and cytokines increased the secretion of IL-6, IL-8, MCP-1, sICAM-1, VEGF and ATP, to significantly higher levels compared to cytokines alone. Peptide5 prevented cytokine release and prevented the increase in ATP release following co-application of HG and cytokines. Adding exogenous ATP negated Peptide5-mediated protection against inflammatory cytokine release in injury conditions. CONCLUSIONS: Our findings show that connexin43 hemichannels play an important role in the amplification and perpetuation of inflammation by mediating an ATP autocrine feedback loop in the inflammasome/inflammation cycle. GENERAL SIGNIFICANCE: Targeting connexin43 hemichannels offers a potential therapeutic strategy to break the inflammatory cycle in diseases such as DR, but also other chronic inflammatory indications.


Subject(s)
Adenosine Triphosphate/metabolism , Connexin 43/physiology , Diabetic Retinopathy/metabolism , Inflammasomes/metabolism , Retinal Pigment Epithelium/metabolism , Adenosine Triphosphate/pharmacology , Autocrine Communication , Cell Line , Connexin 43/antagonists & inhibitors , Cytokines/metabolism , Cytokines/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gap Junctions/physiology , Glucose/pharmacology , Humans , Hyperglycemia/metabolism , Inflammation/metabolism , Retinal Pigment Epithelium/cytology , Vascular Endothelial Growth Factor A/metabolism
7.
Immunol Cell Biol ; 94(10): 955-963, 2016 11.
Article in English | MEDLINE | ID: mdl-27301418

ABSTRACT

The homeostatic chemokine CCL21 has a pivotal role in lymphocyte homing and compartment localisation within the lymph node, and also affects adhesion between immune cells. The effects of CCL21 are modulated by its mode of presentation, with different cellular responses seen for surface-bound and soluble forms. Here we show that plasmin cleaves surface-bound CCL21 to release the C-terminal peptide responsible for CCL21 binding to glycosaminoglycans on the extracellular matrix and cell surfaces, thereby generating the soluble form. Loss of this anchoring peptide enabled the chemotactic activity of CCL21 and reduced cell tethering. Tissue plasminogen activator did not cleave CCL21 directly but enhanced CCL21 processing through generation of plasmin from plasminogen. The tissue plasminogen activator inhibitor neuroserpin prevented processing of CCL21 and blocked the effects of soluble CCL21 on cell migration. Similarly, the plasmin-specific inhibitor α2-antiplasmin inhibited CCL21-mediated migration of human T cells and dendritic cells and tethering of T cells to APCs. We conclude that the plasmin system proteins plasmin, tissue plasminogen activator and neuroserpin regulate CCL21 function in the immune system by controlling the balance of matrix- and cell-bound CCL21.


Subject(s)
Cell Movement/drug effects , Chemokine CCL21/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Plasminogen/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Amino Acid Sequence , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Chemokine CCL21/chemistry , Dendritic Cells/drug effects , Humans , Neuropeptides/pharmacology , Protein Binding/drug effects , Recombinant Proteins/metabolism , Serpins/pharmacology , T-Lymphocytes/drug effects , Tissue Plasminogen Activator/pharmacology , alpha-2-Antiplasmin/pharmacology , Neuroserpin
8.
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.

9.
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
10.
J Neuroinflammation ; 12: 131, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26152369

ABSTRACT

BACKGROUND: The vasculature of the brain is composed of endothelial cells, pericytes and astrocytic processes. The endothelial cells are the critical interface between the blood and the CNS parenchyma and are a critical component of the blood-brain barrier (BBB). These cells are innately programmed to respond to a myriad of inflammatory cytokines or other danger signals. IL-1ß and TNFα are well recognised pro-inflammatory mediators, and here, we provide compelling evidence that they regulate the function and immune response profile of human cerebral microvascular endothelial cells (hCMVECs) differentially. METHODS: We used xCELLigence biosensor technology, which revealed global differences in the endothelial response between IL-1ß and TNFα. xCELLigence is a label-free impedance-based biosensor, which is ideal for acute or long-term comparison of drug effects on cell behaviour. In addition, flow cytometry and multiplex cytokine arrays were used to show differences in the inflammatory responses from the endothelial cells. RESULTS: Extensive cytokine-secretion profiling and cell-surface immune phenotyping confirmed that the immune response of the hCMVEC to IL-1ß was different to that of TNFα. Interestingly, of the 38 cytokines, chemokines and growth factors measured by cytometric bead array, the endothelial cells secreted only 13. Of importance was the observation that the majority of these cytokines were differentially regulated by either IL-1ß or TNFα. Cell-surface expression of ICAM-1 and VCAM-1 were also differentially regulated by IL-1ß or TNFα, where TNFα induced a substantially higher level of expression of both key leukocyte-adhesion molecules. A range of other cell-surface cellular and junctional adhesion molecules were basally expressed by the hCMVEC but were unaffected by IL-1ß or TNFα. CONCLUSIONS: To our knowledge, this is the most comprehensive analysis of the immunological profile of brain endothelial cells and the first direct evidence that human brain endothelial cells are differentially regulated by these two key pro-inflammatory mediators.


Subject(s)
Encephalitis/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Interleukin-1beta/pharmacology , Phenotype , Tumor Necrosis Factor-alpha/pharmacology , Brain/blood supply , Cell Line , Cytokines/metabolism , Encephalitis/metabolism , Endothelial Cells/metabolism , Humans , Immunophenotyping , Intercellular Adhesion Molecule-1/metabolism , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Tight Junction Proteins/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
11.
PLoS One ; 18(10): e0289350, 2023.
Article in English | MEDLINE | ID: mdl-37788259

ABSTRACT

The 'Astrocyte Network' and the understanding of its communication has been posed as a new grand challenge to be investigated by contemporary science. However, communication studies in astrocyte networks have investigated traditional petri-dish in vitro culture models where cells are closely packed and can deviate from the stellate form observed in the brain. Using novel cell patterning approaches, highly organised, regular grid networks of astrocytes on chip, to single-cell fidelity are constructed, permitting a stellate-like in vitro network model to be realised. By stimulating the central cell with a single UV nanosecond laser pulse, the initiation/propagation pathways of stellate-like networks are re-explored. The authors investigate the mechanisms of intercellular Ca2+ communication and discover that stellate-like networks of adult human astrocytes in vitro actually exploit extracellular ATP release as their dominant propagation pathway to cells in the network locally; being observed even down to the nearest neighbour and next nearest neighbouring cells-contrary to the reported gap junction. This discovery has significant ramifications to many neurological conditions such as epilepsy, stroke and aggressive astrocytomas where gap junctions can be targeted. In cases where such gap junction targeting has failed, this new finding suggests that these conditions should be re-visited and the ATP transmission pathway targeted instead.


Subject(s)
Astrocytes , Calcium , Humans , Adult , Astrocytes/metabolism , Calcium/metabolism , Calcium Signaling , Gap Junctions/metabolism , Cell Communication , Communication , Adenosine Triphosphate/metabolism , Cells, Cultured
12.
J Vis Exp ; (199)2023 09 22.
Article in English | MEDLINE | ID: mdl-37811927

ABSTRACT

The blood-brain barrier (BBB) protects the brain parenchyma against harmful pathogens in the blood. The BBB consists of the neurovascular unit, comprising pericytes, astrocytic foot processes, and tightly adhered endothelial cells. Here, the brain endothelial cells form the first line of barrier against blood-borne pathogens. In conditions like cancer and neuroinflammation, circulating factors in the blood can disrupt this barrier. Disease progression significantly worsens post barrier disruption, which permits access to or impairment of regions of the brain. This significantly worsens the prognoses, particularly due to limited treatment options available at the level of the brain. Hence, emerging studies aim to investigate potential therapeutics that can prevent these detrimental factors in the blood from interacting with the brain endothelial cells. The commercially available Electric Cell-Substrate Impedance Sensing (ECIS) and cellZscope instruments measure the impedance across cellular monolayers, such as the BBB endothelium, to determine their barrier strength. Here we detail the use of both biosensors in assessing brain endothelial barrier integrity upon the addition of various stimuli. Crucially, we highlight the importance of their high-throughput capability for concurrent investigation of multiple variables and biological treatments.


Subject(s)
Biosensing Techniques , Neoplasms , Endothelial Cells , Electric Impedance , Cytokines , Brain/blood supply , Blood-Brain Barrier , Pericytes
13.
J Neural Eng ; 20(6)2023 12 11.
Article in English | MEDLINE | ID: mdl-37988746

ABSTRACT

Objective.Glioblastoma (GBM) is the most common and lethal type of high-grade adult brain cancer. The World Health Organization have classed GBM as an incurable disease because standard treatments have yielded little improvement with life-expectancy being 6-15 months after diagnosis. Different approaches are now crucial to discover new knowledge about GBM communication/function in order to establish alternative therapies for such an aggressive adult brain cancer. Calcium (Ca2+) is a fundamental cell molecular messenger employed in GBM being involved in a wide dynamic range of cellular processes. Understanding how the movement of Ca2+behaves and modulates activity in GBM at the single-cell level is relatively unexplored but holds the potential to yield opportunities for new therapeutic strategies and approaches for cancer treatment.Approach.In this article we establish a spatially and temporally precise method for stimulating Ca2+transients in three patient-derived GBM cell-lines (FPW1, RN1, and RKI1) such that Ca2+communication can be studied from single-cell to larger network scales. We demonstrate that this is possible by administering a single optimized ultra-violet (UV) nanosecond laser pulse to trigger GBM Ca2+transients.Main results.We determine that 1.58µJµm-2is the optimal UV nanosecond laser pulse energy density necessary to elicit a single Ca2+transient in the GBM cell-lines whilst maintaining viability, functionality, the ability to be stimulated many times in an experiment, and to trigger further Ca2+communication in a larger network of GBM cells.Significance.Using adult patient-derived mesenchymal GBM brain cancer cell-lines, the most aggressive form of GBM cancer, this work is the first of its kind as it provides a new effective modality of which to stimulate GBM cells at the single-cell level in an accurate, repeatable, and reliable manner; and is a first step toward Ca2+communication in GBM brain cancer cells and their networks being more effectively studied.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Calcium , Cell Line , Brain Neoplasms/drug therapy , Lasers , Cell Line, Tumor
14.
Article in English | MEDLINE | ID: mdl-38083047

ABSTRACT

Glioblastoma (GBM) is a lethal astrocytoma being the most common highest-grade adult brain cancer. GBM tumours are highly invasive and display rapid growth to surrounding areas of the brain. Despite treatment, diagnosed patients continue to have poor prognosis with average survival time of 8 months. Calcium (Ca2+) is a main communication channel used in GBM and its understanding holds the potential to unlock new approaches to treatment. The aim of this work is to provide a first step to accurately evoking Ca2+ transients in GBM cells using single UV nanosecond laser pulses in vitro such that this communication pathway can be more reliably studied from the single-cell to the network level.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioblastoma , Adult , Humans , Glioblastoma/metabolism , Glioblastoma/pathology , Brain/pathology , Brain Neoplasms/pathology , Astrocytoma/pathology , Lasers
15.
Article in English | MEDLINE | ID: mdl-38083627

ABSTRACT

Glioblastoma (GBM) is the most aggressive high-grade brain cancer with a median survival time of <15 months. Due to GBMs fast and infiltrative growth patient prognosis is poor with recurrence after treatment common. Investigating GBMs ability to communicate, specifically via Ca2+ signaling, within its functional tumour networks may unlock new therapeutics to reduce the rapid infiltration and growth which currently makes treatment ineffective. This work aims to produce patterned networks of GBM cells such that the Ca2+ communication at a network level can be repeatedly and reliably investigated.


Subject(s)
Brain Neoplasms , Glioblastoma , Microphysiological Systems , Humans , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Glioblastoma/pathology , Glioblastoma/physiopathology , Silicon
16.
Front Physiol ; 13: 808730, 2022.
Article in English | MEDLINE | ID: mdl-35784870

ABSTRACT

Networks of neurons are typically studied in the field of Criticality. However, the study of astrocyte networks in the brain has been recently lauded to be of equal importance to that of the neural networks. To date criticality assessments have only been performed on networks astrocytes from healthy rats, and astrocytes from cultured dissociated resections of intractable epilepsy. This work, for the first time, presents studies of the critical dynamics and shape collapse of calcium waves observed in cultures of healthy human astrocyte networks in vitro, derived from the human hNT cell line. In this article, we demonstrate that avalanches of spontaneous calcium waves display strong critical dynamics, including power-laws in both the size and duration distributions. In addition, the temporal profiles of avalanches displayed self-similarity, leading to shape collapse of the temporal profiles. These findings are significant as they suggest that cultured networks of healthy human hNT astrocytes self-organize to a critical point, implying that healthy astrocytic networks operate at a critical point to process and transmit information. Furthermore, this work can serve as a point of reference to which other astrocyte criticality studies can be compared.

17.
J Neural Eng ; 18(4)2021 08 24.
Article in English | MEDLINE | ID: mdl-34371484

ABSTRACT

Objective.Platinum nanograss (Ptng) has been demonstrated as an excellent coating to increase the electrode roughness and reduce the impedance of microelectrodes for neural recording. However, the optimisation of the original potentiostatic electrochemical deposition (PSED) method has been performed by the original group only and noin vitrovalidation of functionality was reported.Approach.This study firstly reinvestigates the use of the PSED method for Ptng coating at different charge densities which highlights non-uniformities in the edges of the microelectrodes for increasing deposition charge densities, leading to a decreased impedance which is in fact an artefact. We then introduce a novel Ptng fabrication method of galvanostatic electrochemical deposition (GSED).Main results.We demonstrate that the GSED deposition method also significantly reduces the electrode impedance, raises the charge storage capacity and provides a significantly more planar electrode surface in comparison to the PSED method with negligible edge effects. In addition, we demonstrate how high-quality neural recordings were performed, for the first time, using the Ptng GSED deposition microelectrodes from human hNT neurons and how spiking and bursting were observed.Significance.Thus, the GSED Ptng deposition method presented here provides an alternative method of microelectrode fabrication for neural applications with excellent impedance and planarity of surface.


Subject(s)
Neurons , Platinum , Electric Impedance , Electrochemical Techniques , Humans , Microelectrodes
18.
J Neural Eng ; 18(3)2021 04 17.
Article in English | MEDLINE | ID: mdl-33601342

ABSTRACT

Objective.Cell patterning approaches commonly employed to direct the cytoplasmic outgrowth from cell bodies have been via chemical cues or biomaterial tracks. However, complex network designs using these approaches create problems where multiple tracks lead to manifold obstructions in design. A less common but alternative cell patterning modality is to geometrically design the nodes to project the cytoplasmic processes into a specific direction, thus, removing the need for tracks. Janget alperformed an in-depth study of how rodent neuron primaries could be directed accurately using geometric micro-shapes. In parallel and in contrast, to the work of Janget alwe investigate, for the first time, the effect that micro-shape geometry has on the cytoplasmic process outgrowth of human cells of astrocyte origin using the biomaterial parylene-C.Approach.We investigated eight different types of parylene-C micro-shape on SiO2substrates consisting of the: circle, square, pentagon, hexagon, equilateral triangle and three isosceles triangles with top vertex angles of 14.2°, 28.8°, and 97.6°, respectively. We quantified how each micro-shape influenced the: cell patterning, the directionality of the cytoplasmic process outgrowth and the functionality for human astrocyte.Main results.Human astrocytes became equally well patterned on all different micro-shapes. Human astrocytes could discriminate the underlying micro-shape geometry and preferentially extended processes from the vertices of equilateral triangles and isosceles triangles where the vertex angle equal to 28.8° in a repeatable manner whilst remaining functional.Significance.We demonstrate how human astrocytes are extremely effective at directing their cytoplasmic process outgrowth from the vertices of geometric micro-shapes, in particular the top vertex of triangular shapes. The significance of this work is that it demonstrates that geometric micro-shapes offer an alternative patterning modality to direct cytoplasmic process outgrowth for human astrocytes, which can serve to simplify complex network design, thus, removing the need for tracks.


Subject(s)
Astrocytes , Silicon Dioxide , Biocompatible Materials , Humans , Neurons
19.
Biosensors (Basel) ; 11(5)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069959

ABSTRACT

Electric Cell-Substrate Impedance Sensing (ECIS), xCELLigence and cellZscope are commercially available instruments that measure the impedance of cellular monolayers. Despite widespread use of these systems individually, direct comparisons between these platforms have not been published. To compare these instruments, the responses of human brain endothelial monolayers to TNFα and IL1ß were measured on all three platforms simultaneously. All instruments detected transient changes in impedance in response to the cytokines, although the response magnitude varied, with ECIS being the most sensitive. ECIS and cellZscope were also able to attribute responses to particular endothelial barrier components by modelling the multifrequency impedance data acquired by these instruments; in contrast the limited frequency xCELLigence data cannot be modelled. Consistent with its superior impedance sensing, ECIS exhibited a greater capacity than cellZscope to distinguish between subtle changes in modelled endothelial monolayer properties. The reduced resolving ability of the cellZscope platform may be due to its electrode configuration, which is necessary to allow access to the basolateral compartment, an important advantage of this instrument. Collectively, this work demonstrates that instruments must be carefully selected to ensure they are appropriate for the experimental questions being asked when assessing endothelial barrier properties.


Subject(s)
Biosensing Techniques , Endothelial Cells/physiology , Interleukin-1beta/chemistry , Tumor Necrosis Factor-alpha/chemistry , Electric Impedance , Humans
20.
Annu Int Conf IEEE Eng Med Biol Soc ; 2020: 2245-2248, 2020 07.
Article in English | MEDLINE | ID: mdl-33018454

ABSTRACT

Recently, the study of communication in an 'Astrocyte Network' has been suggested to be of equal importance to that of the traditional 'Neural Network'. In this paper, for the first time, we use nanosecond laser stimulation to stimulate the central cell in an organized grid network of connected human astrocytes in order to observe calcium wave propagation at the single-cell level. We show that the calcium waves indeed propagate from the central astrocyte to the outer periphery of the organized astrocyte network. We observe also, like astrocytes in standard in vitro petri dishes, that the calcium wave propagates through specific connections to the outer periphery of cells rather than in a uniform radial manner predicted by mathematical theory. The results show that such a platform provides an excellent environment to perform repeatable, controlled studies of calcium wave signal propagation through an organized grid network of human astrocytes at single-cell resolution.


Subject(s)
Astrocytes , Calcium Signaling , Astrocytes/metabolism , Calcium/metabolism , Humans , Lasers
SELECTION OF CITATIONS
SEARCH DETAIL