Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neurotrauma ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38468502

ABSTRACT

Cerebral microdialysis (CMD) catheters allow continuous monitoring of patients' cerebral metabolism in severe traumatic brain injury (TBI). The catheters consist of a terminal semi-permeable membrane that is inserted into the brain's interstitium to allow perfusion fluid to equalize with the surrounding cerebral extracellular environment before being recovered through a central non-porous channel. However, it is unclear how far recovered fluid and suspended metabolites have diffused from within the brain, and therefore what volume or region of brain tissue the analyses of metabolism represent. We assessed diffusion of the small magnetic resonance (MR)-detectible molecule gadobutrol from microdialysis catheters in six subjects (complete data five subjects, incomplete data one subject) who had sustained a severe TBI. Diffusion pattern and distance in cerebral white matter were assessed using T1 (time for MR spin-lattice relaxation) maps at 1 mm isotropic resolution in a 3 Tesla MR scanner. Gadobutrol at 10 mmol/L diffused from cerebral microdialysis catheters in a uniform spheroidal (ellipsoid of revolution) pattern around the catheters' semipermeable membranes, and across gray matter-white matter boundaries. Evidence of gadobutrol diffusion was found up to a mean of 13.4 ± 0.5 mm (mean ± standard deviation [SD]) from catheters, but with a steep concentration drop off so that ≤50% of maximum concentration was achieved at ∼4 mm, and ≤10% of maximum was found beyond ∼7 mm from the catheters. There was little variation between subjects. The relaxivity of gadobutrol in human cerebral white matter was estimated to be 1.61 ± 0.38 L.mmol-1sec-1 (mean ± SD); assuming gadobutrol remained extracellular thereby occupying 20% of total tissue volume (interstitium), and concentration equilibrium with perfusion fluid was achieved immediately adjacent to catheters after 24 h of perfusion. No statistically significant change was found in the concentration of the extracellular metabolites glucose, lactate, pyruvate, nor the lactate/pyruvate ratio during gadobutrol perfusion when compared with period of baseline microdialysis perfusion. Cerebral microdialysis allows continuous monitoring of regional cerebral metabolism-the volume of which is now clearer from this study. It also has the potential to deliver small molecule therapies to focal pathologies of the human brain. This study provides a platform for future development of new catheters optimally designed to treat such conditions.

2.
Front Radiol ; 4: 1085834, 2024.
Article in English | MEDLINE | ID: mdl-38356693

ABSTRACT

Rationale and objectives: Cerebral microdialysis is a technique that enables monitoring of the neurochemistry of patients with significant acquired brain injury, such as traumatic brain injury (TBI) and subarachnoid haemorrhage (SAH). Cerebral microdialysis can also be used to characterise the neuro-pharmacokinetics of small-molecule study substrates using retrodialysis/retromicrodialysis. However, challenges remain: (i) lack of a simple, stable, and inexpensive brain tissue model for the study of drug neuropharmacology; and (ii) it is unclear how far small study-molecules administered via retrodialysis diffuse within the human brain. Materials and methods: Here, we studied the radial diffusion distance of small-molecule gadolinium-DTPA from microdialysis catheters in a newly developed, simple, stable, inexpensive brain tissue model as a precursor for in-vivo studies. Brain tissue models consisting of 0.65% weight/volume agarose gel in two kinds of buffers were created. The distribution of a paramagnetic contrast agent gadolinium-DTPA (Gd-DTPA) perfusion from microdialysis catheters using magnetic resonance imaging (MRI) was characterized as a surrogate for other small-molecule study substrates. Results: We found the mean radial diffusion distance of Gd-DTPA to be 18.5 mm after 24 h (p < 0.0001). Conclusion: Our brain tissue model provides avenues for further tests and research into infusion studies using cerebral microdialysis, and consequently effective focal drug delivery for patients with TBI and other brain disorders.

3.
Mol Immunol ; 166: 16-28, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181455

ABSTRACT

Over 500 million people worldwide are affected by diabetes mellitus, a chronic disease that leads to high blood glucose levels and causes severe side effects. The predominant biological marker for diagnosis of diabetes is glycated haemoglobin (GHb). In human blood the predominant reducing sugar, glucose, irreversibly conjugates onto accessible amine groups within Hb. Most methods for diagnosis and monitoring of diabetes selectively detect N-terminal glycation at Val-1 on the ß-globin chain, but not glycation at other sites. Detection of other glycated epitopes of GHb has the potential to provide new information on the extent, duration and timing of elevated glucose, facilitating personalised diagnosis and intelligent diabetic control. In this work, a new anti-GHb Fab antibody (Fab-1) specific for haemoglobin A1c (HbA1c) with nanomolar affinity was discovered via epitope-directed immunisation and phage display. A single chain variable fragment (scFv) antibody derived from Fab-1 retained affinity and specificity for HbA1c, and affinity was enhanced tenfold upon addition of an enhanced green fluorescent protein tag. Both the scFv and Fab-1 recognised an epitope within HbA1c that was distinct from ß-Val-1, and our data suggest that this epitope may include glycation at Lys-66 in the ß-globin chain. To our knowledge, this is the first report of an scFv/Fab anti-glycated epitope antibody that recognises a non-A1c epitope in GHb, and confirms that fructosamine attached to different, discrete glycation sites within the same protein can be resolved from one another by immunoassay.


Subject(s)
Diabetes Mellitus , Single-Chain Antibodies , Sodium Oxybate , Humans , Glycated Hemoglobin , Epitopes , Glucose , beta-Globins
4.
J Cereb Blood Flow Metab ; 43(10): 1685-1701, 2023 10.
Article in English | MEDLINE | ID: mdl-37157814

ABSTRACT

How to optimise glucose metabolism in the traumatised human brain remains unclear, including whether injured brain can metabolise additional glucose when supplied. We studied the effect of microdialysis-delivered 1,2-13C2 glucose at 4 and 8 mmol/L on brain extracellular chemistry using bedside ISCUSflex, and the fate of the 13C label in the 8 mmol/L group using high-resolution NMR of recovered microdialysates, in 20 patients. Compared with unsupplemented perfusion, 4 mmol/L glucose increased extracellular concentrations of pyruvate (17%, p = 0.04) and lactate (19%, p = 0.01), with a small increase in lactate/pyruvate ratio (5%, p = 0.007). Perfusion with 8 mmol/L glucose did not significantly influence extracellular chemistry measured with ISCUSflex, compared to unsupplemented perfusion. These extracellular chemistry changes appeared influenced by the underlying metabolic states of patients' traumatised brains, and the presence of relative neuroglycopaenia. Despite abundant 13C glucose supplementation, NMR revealed only 16.7% 13C enrichment of recovered extracellular lactate; the majority being glycolytic in origin. Furthermore, no 13C enrichment of TCA cycle-derived extracellular glutamine was detected. These findings indicate that a large proportion of extracellular lactate does not originate from local glucose metabolism, and taken together with our earlier studies, suggest that extracellular lactate is an important transitional step in the brain's production of glutamine.


Subject(s)
Glucose , Glutamine , Humans , Glucose/metabolism , Glutamine/metabolism , Brain/metabolism , Microdialysis , Lactic Acid/metabolism , Pyruvic Acid/metabolism , Dietary Supplements
5.
Front Neurol ; 13: 952308, 2022.
Article in English | MEDLINE | ID: mdl-36158966

ABSTRACT

Background: Chronic subdural hematoma (CSDH) is a collection of blood and fluid that arises on the brain surface due to a combination of trauma and/or inflammation. The mainstay of treatment is surgical drainage, but CSDH can recur. Dexamethasone has been shown to reduce CSDH recurrence, but its mechanism of action has not been fully elucidated. Understanding the inflammatory mediators driving CSDH formation and recurrence and how dexamethasone alters this can help develop new therapeutic strategies. Methods: A subgroup of adult patients recruited to the Dex-CSDH trial, randomized to dexamethasone or placebo, who had surgery for their CSDH, were included. CSDH fluid and peripheral blood were collected intraoperatively, from post-operative drains and operated recurrences. Samples were analyzed using a 12-plex panel of inflammatory mediators. Clinical patient data were also reviewed. Results: A total of 52 patients, with a mean age of 76 years, were included. Five recurrent CSDHs occurred. Vascular endothelial growth factor (VEGF) had the highest concentration across all CSDHs, and only matrix metalloproteinase (MMP)-9 had lower concentrations in CSDH compared to plasma but was increased in recurrent CSDHs. The interleukin (IL)-10 concentration was significantly lower in primary CSDHs that recurred. Most inflammatory mediators increased post-operatively, and dexamethasone significantly reduced the post-operative peak in VEGF on day 2, compared to placebo. Conclusion: It is evident that VEGF plays a critical role in the inflammatory response in CSDH. The post-operative reduction with dexamethasone could signal the mechanism by which it reduces recurrence. Novel therapies with a better side-effect profile than dexamethasone should be targeted at VEGF or potential alternatives such as IL-10 supplementation.

6.
Anal Chem ; 93(35): 11929-11936, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34432431

ABSTRACT

The brains of patients suffering from traumatic brain-injury (TBI) undergo dynamic chemical changes in the days following the initial trauma. Accurate and timely monitoring of these changes is of paramount importance for improved patient outcome. Conventional brain-chemistry monitoring is performed off-line by collecting and manually transferring microdialysis samples to an enzymatic colorimetric bedside analyzer every hour, which detects and quantifies the molecules of interest. However, off-line, hourly monitoring means that any subhourly neurochemical changes, which may be detrimental to patients, go unseen and thus untreated. Mid-infrared (mid-IR) spectroscopy allows rapid, reagent-free, molecular fingerprinting of liquid samples, and can be easily integrated with microfluidics. We used mid-IR transmission spectroscopy to analyze glucose, lactate, and pyruvate, three relevant brain metabolites, in the extracellular brain fluid of two TBI patients, sampled via microdialysis. Detection limits of 0.5, 0.2, and 0.1 mM were achieved for pure glucose, lactate, and pyruvate, respectively, in perfusion fluid using an external cavity-quantum cascade laser (EC-QCL) system with an integrated transmission flow-cell. Microdialysates were collected hourly, then pooled (3-4 h), and measured consecutively using the standard ISCUSflex analyzer and the EC-QCL system. There was a strong correlation between the compound concentrations obtained using the conventional bedside analyzer and the acquired mid-IR absorbance spectra, where a partial-least-squares regression model was implemented to compute concentrations. This study demonstrates the potential utility of mid-IR spectroscopy for continuous, automated, reagent-free, and online monitoring of the dynamic chemical changes in TBI patients, allowing a more timely response to adverse brain metabolism and consequently improving patient outcomes.


Subject(s)
Extracellular Fluid , Lasers, Semiconductor , Glucose , Humans , Microdialysis , Spectrophotometry, Infrared
7.
J Cereb Blood Flow Metab ; 40(1): 67-84, 2020 01.
Article in English | MEDLINE | ID: mdl-30226401

ABSTRACT

Metabolic dysfunction is a key pathophysiological process in the acute phase of traumatic brain injury (TBI). Although changes in brain glucose metabolism and extracellular lactate/pyruvate ratio are well known, it was hitherto unknown whether these translate to downstream changes in ATP metabolism and intracellular pH. We have performed the first clinical voxel-based in vivo phosphorus magnetic resonance spectroscopy (31P MRS) in 13 acute-phase major TBI patients versus 10 healthy controls (HCs), at 3T, focusing on eight central 2.5 × 2.5 × 2.5 cm3 voxels per subject. PCr/γATP ratio (a measure of energy status) in TBI patients was significantly higher (median = 1.09) than that of HCs (median = 0.93) (p < 0.0001), due to changes in both PCr and ATP. There was no significant difference in PCr/γATP between TBI patients with favourable and unfavourable outcome. Cerebral intracellular pH of TBI patients was significantly higher (median = 7.04) than that of HCs (median = 7.00) (p = 0.04). Alkalosis was limited to patients with unfavourable outcome (median = 7.07) (p < 0.0001). These changes persisted after excluding voxels with > 5% radiologically visible injury. This is the first clinical demonstration of brain alkalosis and elevated PCr/γATP ratio acutely after major TBI. 31P MRS has potential for non-invasively assessing brain injury in the absence of structural injury, predicting outcome and monitoring therapy response.


Subject(s)
Brain Injuries, Traumatic/metabolism , Magnetic Resonance Imaging/methods , Phosphorus , Adenosine Triphosphate/metabolism , Adult , Alkalosis/diagnostic imaging , Brain Injuries, Traumatic/diagnostic imaging , Case-Control Studies , Energy Metabolism , Female , Humans , Male , Middle Aged , Prognosis
8.
J Neurotrauma ; 37(1): 106-114, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31298609

ABSTRACT

Cerebral microdialysis (CMD) is used in severe traumatic brain injury (TBI) in order to recover metabolites in brain extracellular fluid (ECF). To recover larger proteins and avoid fluid loss, albumin supplemented perfusion fluid (PF) has been utilized, but because of regulatory changes in the European Union, this is no longer practicable. The aim with this study was to see whether fluid, absolute (AR), and relative (RR) recovery for the novel carrier, Dextran 500, was better than conventional PF for a range of cytokines and chemokines. An in vitro setup mimicking conditions observed in the neurocritical care of TBI patients was used, utilizing 100-kDa molecular-weight cut-off CMD catheters inserted through a triple-lumen bolt cranial access device into an external solution with diluted cytokine standards in known concentrations for 48 h (divided into 6-h epochs). Samples were run on a 39-plex Luminex (Luminex Corporation, Austin, TX) assay to assess cytokine concentrations. We found that fluid recovery was inadequate in 50% of epochs with conventional PF, whereas Dextran PF overcame this limitation. The AR was higher in the Dextran PF samples for a majority of cytokines, and RR was significantly increased for macrophage colony-stimulating factor and transforming growth factor-alpha. In summary, Dextran PF improved fluid and cytokine recovery as compared to conventional PF and is a suitable alternative to albumin supplemented PF for protein microdialysis.


Subject(s)
Biomarkers/analysis , Brain Injuries, Traumatic/metabolism , Cytokines/analysis , Dextrans , Microdialysis/methods , Extracellular Fluid/metabolism , Humans , In Vitro Techniques , Inflammation/etiology
9.
J Neurotrauma ; 37(1): 93-105, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31452443

ABSTRACT

Neuroinflammation has been shown to mediate the pathophysiological response following traumatic brain injury (TBI). Accumulating evidence implicates astrocytes as key immune cells within the central nervous system (CNS), displaying both pro- and anti-inflammatory properties. The aim of this study was to investigate how in vitro human astrocyte cultures respond to cytokines across a concentration range that approximates the aftermath of human TBI. To this end, enriched cultures of human induced pluripotent stem cell (iPSC)-derived astrocytes were exposed to interleukin-1ß (IL-1ß) (1-10,000 pg/mL), IL-4 (1-10,000 pg/mL), IL-6 (100-1,000,000 pg/mL), IL-10 (1-10,000 pg/mL) and tumor necrosis factor (TNF)-α (1-10,000 pg/mL). After 1, 24, 48 and 72 h, cultures were fixed and immunolabeled, and the secretome/supernatant was analyzed at 24, 48, and 72 h using a human cytokine/chemokine 39-plex Luminex assay. Data were compared to previous in vitro studies of neuronal cultures and clinical TBI studies. The secretome revealed concentration-, time- and/or both concentration- and time-dependent production of downstream cytokines (29, 21, and 17 cytokines, respectively, p<0.05). IL-1ß exposure generated the most profound downstream response (27 cytokines), IL-6 and TNF had intermediate responses (13 and 11 cytokines, respectively), whereas IL-4 and IL-10 only led to weak responses over time or in escalating concentration (8 and 8 cytokines, respectively). Notably, expression of IL-1ß, IL-6, and TNF cytokine receptor mRNA was higher in astrocyte cultures than in neuronal cultures. Several secreted cytokines had temporal trajectories, which corresponded to those seen in the aftermath of human TBI. In summary, iPSC-derived astrocyte cultures exposed to cytokine concentrations reflecting those in TBI generated an increased downstream cytokine production, particularly IL-1ß. Although more work is needed to better understand how different cells in the CNS respond to the neuroinflammatory milieu after TBI, our data shows that iPSC-derived astrocytes represent a tractable model to study cytokine stimulation in a cell type-specific manner.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Cytokines/metabolism , Cytokines/pharmacology , Astrocytes/drug effects , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism
10.
Mikrochim Acta ; 187(1): 13, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31802241

ABSTRACT

A proof-of-concept aptamer-based optical assay is described for the determination of the immuno signalling molecule interleukin-6 (IL-6), a key marker of acute inflammation. The optical assay is based on the aggregation of gold nanoparticles (AuNP) coated in two complimentary "sandwich-style" aptamers, each with different IL-6 target moieties. IL-6 will recognise the complimentary aptamer pair and bind to it, thereby causing the aggregation of the corresponding functionalised nanoparticles. The aggregation of the AuNPs after exposure to IL-6 induces a visible colour change from red to pink, with a corresponding change in the absorption maximum from 520 to 540 nm. The change in the absorption maximum can be monitored visually, or by using a spectrophotometer or a plate reader. The optimal size and functionalisation of aptamer-coated AuNPs, and the potential assay formats were investigated using UV-vis spectrophotometry, transmission electron microscopy, and dynamic light scattering. The optical assay was applied for detecting mouse IL-6 in a mixed protein solution as a representative biological sample. The assay works in the 3.3 to 125 µg·mL-1 IL-6 concentration range, and the detection limit (at S/N = 3) is 1.95 µg·mL-1. This study was performed as a proof-of-concept demonstration of this versatile assay design, with a view to developing a similar assay for use in clinical samples in future. Graphical abstractSchematic representation of the aggregation of aptamer-functionalised nanoparticles in the presence of interleukin-6 (IL-6). The presence of mouse IL-6 in a mixed protein solution leads to a visible colour change, and a change in the absorption spectrum of the nanoparticles.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Gold/chemistry , Interleukin-6/analysis , Metal Nanoparticles/chemistry , Aptamers, Nucleotide/metabolism , Inflammation/diagnosis , Interleukin-6/metabolism , Kinetics
11.
Expert Rev Neurother ; 19(3): 227-242, 2019 03.
Article in English | MEDLINE | ID: mdl-30848963

ABSTRACT

INTRODUCTION: Traumatic Brain Injury (TBI) is a leading cause of death and disability in young people, affecting 69 million people annually, worldwide. The initial trauma disrupts brain homeostasis resulting in metabolic dysfunction and an inflammatory cascade, which can then promote further neurodegenerative effects for months or years, as a 'secondary' injury. Effective targeting of the cerebral inflammatory system is challenging due to its complex, pleiotropic nature. Cell metabolism plays a key role in many diseases, and increased disturbance in the TBI metabolic state is associated with poorer patient outcomes. Investigating critical metabolic pathways, and their links to inflammation, can potentially identify supplements which alter the brain's long-term response to TBI and improve recovery. Areas covered: The authors provide an overview of literature on metabolism and inflammation following TBI, and from relevant pre-clinical and clinical studies, propose therapeutic strategies. Expert opinion: There is still no specific active drug treatment for TBI. Changes in metabolic and inflammatory states have been reported after TBI and appear linked. Understanding more about abnormal cerebral metabolism following TBI, and its relationship with cerebral inflammation, will provide essential information for designing therapies, with implications for neurocritical care and for alleviating long-term disability and neurodegeneration in post-TBI patients.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/physiopathology , Brain/metabolism , Inflammation/physiopathology , Anti-Inflammatory Agents/therapeutic use , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Humans , Inflammation/etiology , Young Adult
12.
Sci Rep ; 8(1): 11140, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30042490

ABSTRACT

A key pathophysiological process and therapeutic target in the critical early post-injury period of traumatic brain injury (TBI) is cell mitochondrial dysfunction; characterised by elevation of brain lactate/pyruvate (L/P) ratio in the absence of hypoxia. We previously showed that succinate can improve brain extracellular chemistry in acute TBI, but it was not clear if this translates to a change in downstream energy metabolism. We studied the effect of microdialysis-delivered succinate on brain energy state (phosphocreatine/ATP ratio (PCr/ATP)) with 31P MRS at 3T, and tissue NADH/NAD+ redox state using microdialysis (L/P ratio) in eight patients with acute major TBI (mean 7 days). Succinate perfusion was associated with increased extracellular pyruvate (+26%, p < 0.0001) and decreased L/P ratio (-13%, p < 0.0001) in patients overall (baseline-vs-supplementation over time), but no clear-cut change in 31P MRS PCr/ATP existed in our cohort (p > 0.4, supplemented-voxel-vs-contralateral voxel). However, the percentage decrease in L/P ratio for each patient following succinate perfusion correlated significantly with their percentage increase in PCr/ATP ratio (Spearman's rank correlation, r = -0.86, p = 0.024). Our findings support the interpretation that L/P ratio is linked to brain energy state, and that succinate may support brain energy metabolism in select TBI patients suffering from mitochondrial dysfunction.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Energy Metabolism/drug effects , NAD/metabolism , Phosphates/metabolism , Succinic Acid/pharmacology , Adenosine Triphosphate/metabolism , Adult , Aged , Brain/metabolism , Brain Chemistry/drug effects , Female , Humans , Hydrogen-Ion Concentration/drug effects , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Microdialysis/methods , Middle Aged , Oxidation-Reduction , Perfusion , Phosphocreatine/metabolism , Pilot Projects , Prospective Studies , Pyruvic Acid/metabolism , Signal Transduction/drug effects , Statistics, Nonparametric , Succinic Acid/administration & dosage , Succinic Acid/metabolism , Treatment Outcome , Young Adult
13.
Anal Bioanal Chem ; 409(21): 5031-5042, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28664332

ABSTRACT

Cerebral microdialysis is a sampling technique which offers much potential for understanding inflammatory pathophysiology following traumatic brain injury (TBI). At present, the recovery of cytokines via microdialysis in clinical studies is not straightforward primarily due to their size, steric properties and low concentrations. Heparin and heparin-coated microspheres have previously shown promise as cytokine-binding agents for enhanced microdialysis sampling in animal models (Duo and Stenken in Anal Bioanal Chem 399(2):773-82, 2011; Anal Bioanal Chem 399(2):783-93, 2011). However, there are several factors limiting application for microdialysis in patients. The aim of this study was to produce heparin-coated gold nanoparticles as cytokine capture agents for enhanced microdialysis sampling, potentially applicable to a clinical setting. Gold nanoparticles (AuNP) were chemically conjugated to heparin via a bifunctional polyethylene glycol (PEG) linker. The heparin-AuNP (AuNP-Hep) were characterised, demonstrating the successful addition of heparin to the gold surface. The performance of the AuNP-Hep during in vitro testing was compared both to current methodology (Helmy et al. in J Neurotrauma 26(4):549-61, 2009) and to the heparin-coated microspheres developed by Duo and Stenken (Anal Bioanal Chem 399(2):773-82, 2011; Anal Bioanal Chem 399(2):783-93, 2011). The AuNP-Hep yielded a higher recovery of cytokines compared to current methodology and heparin-coated microspheres, during in vitro testing designed to mimic the human environment and the intensive care unit. In this study, AuNP-Hep were developed for enhanced microdialysis sampling of cytokines, potentially applicable in a clinical setting. Based on the success of the AuNP-Hep in vitro, the proposed method offers an alternative to the use of current protocols that rely on a blood product (albumin) for microdialysis sampling of cytokines in patients.


Subject(s)
Gold/chemistry , Heparin/chemistry , Metal Nanoparticles/chemistry , Microdialysis/methods , Cytokines/chemistry , Humans , In Vitro Techniques , Microscopy, Electron, Transmission
14.
J Neuroinflammation ; 14(1): 108, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28558815

ABSTRACT

Chronic subdural haematoma (CSDH) is an encapsulated collection of blood and fluid on the surface of the brain. Historically considered a result of head trauma, recent evidence suggests there are more complex processes involved. Trauma may be absent or very minor and does not explain the progressive, chronic course of the condition. This review focuses on several key processes involved in CSDH development: angiogenesis, fibrinolysis and inflammation. The characteristic membrane surrounding the CSDH has been identified as a source of fluid exudation and haemorrhage. Angiogenic stimuli lead to the creation of fragile blood vessels within membrane walls, whilst fibrinolytic processes prevent clot formation resulting in continued haemorrhage. An abundance of inflammatory cells and markers have been identified within the membranes and subdural fluid and are likely to contribute to propagating an inflammatory response which stimulates ongoing membrane growth and fluid accumulation. Currently, the mainstay of treatment for CSDH is surgical drainage, which has associated risks of recurrence requiring repeat surgery. Understanding of the underlying pathophysiological processes has been applied to developing potential drug treatments. Ongoing research is needed to identify if these therapies are successful in controlling the inflammatory and angiogenic disease processes leading to control and resolution of CSDH.


Subject(s)
Hematoma, Subdural, Chronic/complications , Inflammation/drug therapy , Inflammation/etiology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/etiology , Animals , Hematoma, Subdural, Chronic/drug therapy , Humans
15.
Sci Rep ; 7(1): 1003, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28432362

ABSTRACT

Mitochondrial dysfunction, the inability to efficiently utilise metabolic fuels and oxygen, contributes to pathological changes following traumatic spinal cord or traumatic brain injury (TBI). In the present study, we tested the hypothesis that succinate supplementation can improve cellular energy state under metabolically stressed conditions in a robust, reductionist in vitro model of mitochondrial dysfunction in which primary mixed glial cultures (astrocytes, microglia and oligodendrocytes) were exposed to the mitochondrial complex I inhibitor rotenone. Cellular response was determined by measuring intracellular ATP, extracellular metabolites (glucose, lactate, pyruvate), and oxygen consumption rate (OCR). Rotenone produced no significant changes in glial ATP levels. However, it induced metabolic deficits as evidenced by lactate/pyruvate ratio (LPR) elevation (a clinically-established biomarker for poor outcome in TBI) and decrease in OCR. Succinate addition partially ameliorated these metabolic deficits. We conclude that succinate can improve glial oxidative metabolism, consistent our previous findings in TBI patients' brains. The mixed glial cellular model may be useful in developing therapeutic strategies for conditions involving mitochondrial dysfunction, such as TBI.


Subject(s)
Energy Metabolism/drug effects , Mitochondria/drug effects , Neuroglia/cytology , Rotenone/adverse effects , Succinic Acid/pharmacology , Animals , Brain Injuries, Traumatic/diet therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dietary Supplements , Mitochondria/physiology , Models, Biological , Neuroglia/drug effects , Neuroglia/metabolism , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/diet therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...