Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1783 2022-05-09 19:20:09 |
2 format change Meta information modification 1783 2022-05-10 03:58:17 | |
3 format change Meta information modification 1783 2022-05-11 08:09:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zimphango, C.; Alimagham, F.; , . Monitoring Neurochemistry in Traumatic Brain Injury Patients. Encyclopedia. Available online: https://encyclopedia.pub/entry/22724 (accessed on 27 July 2024).
Zimphango C, Alimagham F,  . Monitoring Neurochemistry in Traumatic Brain Injury Patients. Encyclopedia. Available at: https://encyclopedia.pub/entry/22724. Accessed July 27, 2024.
Zimphango, Chisomo, Farah Alimagham,  . "Monitoring Neurochemistry in Traumatic Brain Injury Patients" Encyclopedia, https://encyclopedia.pub/entry/22724 (accessed July 27, 2024).
Zimphango, C., Alimagham, F., & , . (2022, May 09). Monitoring Neurochemistry in Traumatic Brain Injury Patients. In Encyclopedia. https://encyclopedia.pub/entry/22724
Zimphango, Chisomo, et al. "Monitoring Neurochemistry in Traumatic Brain Injury Patients." Encyclopedia. Web. 09 May, 2022.
Monitoring Neurochemistry in Traumatic Brain Injury Patients
Edit

In a traumatically injured brain, the cerebral microdialysis technique allows continuous sampling of fluid from the brain’s extracellular space. The retrieved brain fluid contains useful metabolites that indicate the brain’s energy state. Assessment of these metabolites along with other parameters, such as intracranial pressure, brain tissue oxygenation, and cerebral perfusion pressure, may help inform clinical decision making, guide medical treatments, and aid in the prognostication of patient outcomes. Currently, brain metabolites are assayed on bedside analysers and results can only be achieved hourly. This is a major drawback because critical information within each hour is lost. To address this, recent advances have focussed on developing biosensing techniques for integration with microdialysis to achieve continuous online monitoring. 

biosensors traumatic brain injury neurochemistry cerebral microdialysis cerebral metabolism

1. Introduction

Traumatic brain injury (TBI) is a health burden affecting over 50 million people each year globally [1][2]. Its long-term adverse effects to the brain may create a significant burden implicating social integration, independence, and employability. Population-based studies have reported that 238,000 people each year sustain a TBI in the UK, and it is the overall leading cause of death in young adults (25–35 years) in developed countries [3]. In 2013, TBIs in the USA led to about 2.5 million emergency department visits, 282,000 hospitalisations, and 56,000 deaths [4]. Mild or severe TBI patients are at risk of secondary brain injury that arises from complex physiological responses to the initial injury. These complex physiological processes are thought to include a series of sequential events including altered cerebral metabolism. Monitoring cerebral metabolism along with other protocols to identify early changes and prevent secondary brain injuries during neurocritical care management can improve outcomes of severe TBI patients [5][6]. Currently, cerebral metabolism is monitored using cerebral microdialysis, an invasive technique in which interstitial brain fluid can be sampled. The sampled fluid (also termed microdialysate) is usually analysed hourly offline using a bedside analyser, for which its operating principle is based on enzymatic reactions [7][8]. However, the data analysed are retrospective, hindering real-time interventions. Thus, effective and timely measurements of the microdialysates are required to predict the onset of secondary brain damage. To fulfil this, emerging research has integrated microdialysis with biosensors for continuous online detection of relevant cerebral metabolites found in microdialysates. The intent of these technologies is to provide accurate and actionable information that may help inform clinical decision making and aid in the prognostication of patients’ outcome.

2. Importance of Monitoring Brain Metabolism for TBI

2.1. Traumatic Brain Injury (TBI)

TBI is an insult to the brain due to an external force causing damage to the brain’s structure and function [9]. It represents mild, moderate, and severe effects of physical assault to the brain that may cause sequential, primary, or secondary ramifications. Thus, TBI can be classed into a primary mechanical injury, which is non-amenable to medical treatment and a delayed secondary brain injury involving various changes at the cellular and molecular level that could in theory be counteracted. The sudden mechanical injury is a widespread tearing, shearing, or stretching of axons, referred to as axonal injury, as well as contusions, hemorrhages, and lacerations [10]. Delayed secondary injury evolves hours or days after the initial primary mechanical trauma due to neuronal and glial dysfunction, neuroinflammation, cerebral oedema, and metabolic changes. Consequently, this leads to various physiologic alterations including hypoperfusion, blood–brain barrier (BBB) disruption, oxidative injury, and mitochondrial dysfunction [11][12][13][14][15][16]. This complexity of secondary brain injury poses diagnostic and therapeutic challenges; therefore, additional invasive monitoring interventions are required during neurocritical care of severe TBI patients. These additional metrics may provide insights into brain tissue oxygenation, intracranial pressure (ICP), cerebral perfusion pressure (CPP), electrophysiology, and local brain metabolism [11]. Abnormal local brain metabolism is linked to poor patient clinical outcomes, thus interrogating the brain to monitor its chemistry is necessary [17].

2.2. Cerebral Metabolism

Evidence shows that cerebral metabolism is disturbed following TBI, although the exact mechanisms are incompletely understood due to the complex and heterogeneous nature of TBI [7]. The brain uses glucose as a preferred substrate for energy consumption, so the regulation of cerebral glucose metabolism is crucial. Oxidative metabolism of glucose provides most of the ATPs utilised by the brain. However, biosynthetic routes that branch from the glycolytic pathway and the tricarboxylic acid (TCA) cycle and other pathways including the pentose phosphate shunt, glucose storage as glycogen, and the malate-aspartate shuttle all have significant roles [18]. Understanding of the altered cerebral metabolism is incomplete without knowledge of the glucose metabolism in an uninjured brain. Figure 1 presents the major energy pathways in the brain.
Figure 1. Illustration of glucose, lactate, and pyruvate within the major energy pathways. Hexokinases uses ATP to phosphorylate glucose to glucose 6P (glucose 6-phosphate) in the first irreversible step of glycolysis. Glucose 6P can either undergo alternative metabolic fates or continue down the glycolytic pathway to generate pyruvate. Pyruvate can be utilised either by oxidative metabolism via the tricarboxylic acid (TCA) cycle or remain in the cytosol where it is converted to lactate. The glycolytic pathway that takes place in the cytoplasm produces a net yield of 2 ATP per molecule of glucose (2 ATP molecules are utilised early on but then paid back later with the generation of 4 ATP). Overall, the yield per mole of glucose metabolised fully to CO2 by combination of glycolysis and mitochondrial metabolism is theoretically 36–38 moles of ATP.
In the uninjured brain, glucose uptake by both neuronal and glial cells undergoes oxidation via glycolysis to form two pyruvate molecules. Glycolysis is independent of oxygen and, thus, can occur under either aerobic or anaerobic conditions. The energy yield of ATP per molecule of glucose depends on whether mitochondrial shuttle mechanisms are operational or not. To generate one molecule of pyruvate by glycolysis, one molecule of NAD+ is converted into NADH, which must be recycled (oxidised) back to NAD+ if glycolysis is to be sustained. One recycling mechanism is by the electron-transport chains of mitochondria (if operational). As NADH cannot cross the mitochondrial membrane, the requisite hydrogens and electrons are transferred indirectly by “shuttle” mechanisms-malate-aspartate shuttle system and/or the glycerol-3-phosphate shuttle [19]. NADH can also be recycled to NAD+ by the lactate dehydrogenase (LDH)-mediated conversion of pyruvate to lactate in the cytosol, as an epilogue to “anaerobic” glycolysis.
The overall reaction of glycolysis and net yield of ATP molecules is presented in Equation (1).
Pyruvate, the end-product of glycolysis, can enter mitochondria where it is converted to acetyl-CoA by pyruvate dehydrogenase. Acetyl-CoA is further metabolised in the tricarboxylic acid (TCA) cycle in mitochondria. The sum of all the reactions in the TCA cycle is presented in Equation (2).
Subsequently, NADH and FADH2 are utilised by the mitochondrial electron transport chain (by Complexes I and II, respectively); electrons are transferred to complexes III and IV, where O2 is the terminal electron acceptor on Complex IV, followed by ATP synthesis by ATP synthase (also termed Complex V) [20]. The yield per molecule of glucose metabolised fully to CO2 (by combined glycolysis, NADH shuttling, and mitochondrial respiration) is theoretically 36–38 ATP molecules. However, the actual yield is considered somewhat lower [21][22].

2.3. Altered Cerebral Metabolism Due to TBI

Imbalance in cerebral glucose metabolism following TBI is well-documented [7][23][24] and attributable, at least partly, to the altered ATP production in the brain’s major energy pathways. Due to a high energy demand following TBI, abnormally low levels (<0.8 mM) of extracellular glucose occur [17], possibly because of upregulated glucose uptake by neurones and glia. Conversely, neurones and glia may sometimes be too damaged to take up glucose from extracellular fluid, leading to hypometabolism characterised by abnormally high extracellular glucose. Thus, there is an optimum extracellular glucose range, although there is insufficient evidence to define this exactly [25]. Extracellular lactate can also be utilised as an alternative fuel [24]. 13C-labelled microdialysis studies have demonstrated that the traumatically injured brain uses lactate via the TCA cycle [24][26]. Another 13C-labelled microdialysis study found lactate production from 1,2-13C2 glucose via glycolysis and to a lesser extent via PPP [23]. Lactate was also identified as a spin-out product (cataplerosis) from the TCA cycle in 13C-labelled microdialysis studies using 2,3-13C2 succinate as a substrate [11][23].
A persistent high lactate/pyruvate ratio (LPR) (LPR > 25 or >40) indicates metabolic dysfunction or crisis. In a microdialysis study of 233 TBI patients, acute-phase LPR > 25 was associated with poor clinical outcomes 6 months later [17]. High LPR, despite seemingly adequate oxygen and glucose delivery to brain tissues, is regarded as indicating mitochondrial dysfunction [11][27]. The concentrations of lactate and pyruvate and their ratio (LPR) provide useful information about the cellular redox state in the region of interest. The extracellular LPR is thought to reflect the LPR in the cytoplasm—itself in equilibrium with cytoplasmic NADH/NAD+ ratio [28]. Glucose, lactate, pyruvate, and LPR were cited as the most clinically relevant biomarkers in a consensus statement from the 2014 International Microdialysis Forum [25]. Timely assessment of these is, therefore, essential in the early detection of secondary brain injury allowing prompt interventions. Table 1 summarises neuroprotective interventions for altered neurochemistry.
Table 1. Summary of neuroprotective interventions for altered cerebral metabolism [29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44].

3. Review of Sensor Technologies for Brain Metabolism

In selecting scholarly articles to discuss here, emphasis was placed on work published in the areas of biosensors, TBI, cerebral metabolism, and microdialysis in the last 10 years. Table 2 summarises some of the most relevant articles discussed here. Those biosensors are in the research and development phases and are not approved for routine clinical use.
Table 2. A summary comparison of recent key examples of biosensors integrated with microdialysis for brain metabolite monitoring [45][46][47][48][49][50][51][52].

Study

Sensor Type

Setting

Comments

Papadimitriou et al. (2016) [45]

Enzymatic-electrochemical

In-vitro

Measured 0–100 μM glucose concentration, with 25 μM increments, in a microdialysate stream.

Pagkalos et al. (2018) [46]

Enzymatic-electrochemical

In-vitro

Measured 0–50 μM lactate concentrations with 12.5 μM increments using enzymatic based sensor with LoD range 2.5 to 9.5 nM, in a microdialysate stream.

Tageldeen et al. (2020) [47]

Enzymatic-electrochemical

In-vitro

Measured 0–1 mM glucose and lactate, changing concentrations. LoDs of 0.85 and 1.3 μM for glucose and lactate, respectively, in a microdialysate stream.

Robbins et al. (2019) [48]

Enzymatic-electrochemical

In-vivo

(rats)

Reported progressive decrease in glucose in microdialysates from a cortical impact injury.

Rogers et al. (2017) [50]

Enzymatic-electrochemical

In-vivo (human)

Continuous online microdialysis measurements in TBI patients; monitoring duration > 6 h; glucose, lactate, and K+ levels in spreading depolarisation (K+ was measured by an ion-selective electrode).

Gowers et al. (2019) [52]

Enzymatic-electrochemical

In-vivo (human)

Detected a sudden surge of lactate levels during continuous online dialysate measurements in TBI patients.

Gifford et al. (2021) [51]

Enzymatic-electrochemical

In-vivo (human)

Reported declining glucose levels in 3 TBI patients, and persistent low glucose in 1 TBI patient, in dexamethasone-enhanced continuous online microdialysis.

Alimagham et al. (2021) [49]

Optical

(mid-IR)

Ex-vivo (human)

Microdialysate measurements from TBI patients, offline. LoDs of 0.5, 0.2, and 0.1 mM for glucose, lactate, and pyruvate respectively. Quantification of brain metabolites was compared with a conventional enzymatic-colorimetric microdialysis analyser (ISCUSflex).

References

  1. Maas, A.I.R.; Menon, D.K.; Adelson, P.D.; Andelic, N.; Bell, M.J.; Belli, A.; Bragge, P.; Brazinova, A.; Büki, A.; Chesnut, R.M.; et al. Traumatic brain injury: Integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017, 16, 987–1048.
  2. Dewan, M.C.; Rattani, A.; Gupta, S.; Baticulon, R.E.; Hung, Y.C.; Punchak, M.; Agrawal, A.; Adeleye, A.O.; Shrime, M.G.; Rubiano, A.M.; et al. Estimating the global incidence of traumatic brain injury. J. Neurosurg. 2018, 130, 1080–1097.
  3. Barlow, K.M. Traumatic brain injury. Handb. Clin. Neurol. 2013, 112, 891–904.
  4. Taylor, C.A.; Bell, J.M.; Breiding, M.J.; Xu, L. Traumatic Brain Injury-Related Emergency Department Visits, Hospitalizations, and Deaths-United States, 2007 and 2013. MMWR Surveill. Summ. 2017, 66, 1–16.
  5. Vidgeon, S.D.; Strong, A.J. Multimodal Cerebral Monitoring in Traumatic Brain Injury. J. Intensive Care Soc. 2011, 12, 126–133.
  6. Wijayatilake, D.S.; Shepherd, S.J. What’s new in the management of traumatic brain injury on neuro ICU? Curr. Opin. Anaesthesiol. 2014, 27, 459–464.
  7. Carpenter, K.L.; Jalloh, I.; Gallagher, C.N.; Grice, P.; Howe, D.J.; Mason, A.; Timofeev, I.; Helmy, A.; Murphy, M.P.; Menon, D.K.; et al. 13C-labelled microdialysis studies of cerebral metabolism in TBI patients. Eur. J. Pharm. Sci. 2014, 57, 87–97.
  8. Helmy, A.; Carpenter, K.L.; Hutchinson, P.J. Microdialysis in the human brain and its potential role in the development and clinical assessment of drugs. Curr. Med. Chem. 2007, 14, 1525–1537.
  9. Menon, D.K.; Schwab, K.; Wright, D.W.; Maas, A.I. Position statement: Definition of traumatic brain injury. Arch. Phys. Med. Rehabil. 2010, 91, 1637–1640.
  10. Ng, S.Y.; Lee, A.Y.W. Traumatic Brain Injuries: Pathophysiology and Potential Therapeutic Targets. Front. Cell. Neurosci. 2019, 13, 528.
  11. Khellaf, A.; Khan, D.Z.; Helmy, A. Recent advances in traumatic brain injury. J. Neurol. 2019, 266, 2878–2889.
  12. Chamoun, R.; Suki, D.; Gopinath, S.P.; Goodman, J.C.; Robertson, C. Role of extracellular glutamate measured by cerebral microdialysis in severe traumatic brain injury. J. Neurosurg. 2010, 113, 564–570.
  13. Dyhrfort, P.; Shen, Q.; Clausen, F.; Thulin, M.; Enblad, P.; Kamali-Moghaddam, M.; Lewén, A.; Hillered, L. Monitoring of Protein Biomarkers of Inflammation in Human Traumatic Brain Injury Using Microdialysis and Proximity Extension Assay Technology in Neurointensive Care. J. Neurotrauma 2019, 36, 2872–2885.
  14. Lassarén, P.; Lindblad, C.; Frostell, A.; Carpenter, K.L.H.; Guilfoyle, M.R.; Hutchinson, P.J.A.; Helmy, A.; Thelin, E.P. Systemic inflammation alters the neuroinflammatory response: A prospective clinical trial in traumatic brain injury. J. Neuroinflamm. 2021, 18, 221.
  15. Di, X.; Lyeth, B.G.; Hamm, R.J.; Bullock, M.R. Voltage-dependent Na+/K+ ion channel blockade fails to ameliorate behavioral deficits after traumatic brain injury in the rat. J. Neurotrauma 1996, 13, 497–504.
  16. Guilfoyle, M.R.; Carpenter, K.L.; Helmy, A.; Pickard, J.D.; Menon, D.K.; Hutchinson, P.J. Matrix Metalloproteinase Expression in Contusional Traumatic Brain Injury: A Paired Microdialysis Study. J. Neurotrauma 2015, 32, 1553–1559.
  17. Timofeev, I.; Carpenter, K.L.H.; Nortje, J.; Al-Rawi, P.G.; O’Connell, M.T.; Czosnyka, M.; Smielewski, P.; Pickard, J.D.; Menon, D.K.; Kirkpatrick, P.J.; et al. Cerebral extracellular chemistry and outcome following traumatic brain injury: A microdialysis study of 223 patients. Brain 2011, 134, 484–494.
  18. Dienel, G.A. Brain Glucose Metabolism: Integration of Energetics with Function. Physiol. Rev. 2019, 99, 949–1045.
  19. McKenna, M.C.; Waagepetersen, H.S.; Schousboe, A.; Sonnewald, U. Neuronal and astrocytic shuttle mechanisms for cytosolic-mitochondrial transfer of reducing equivalents: Current evidence and pharmacological tools. Biochem. Pharm. 2006, 71, 399–407.
  20. Rich, P.R.; Maréchal, A. The mitochondrial respiratory chain. Essays Biochem. 2010, 47, 1–23.
  21. Berg, J.M.; Tymoczko, J.L.; Gatto, G.J., Jr.; Stryer, L. Biochemistry, 9th ed.; McMillan Learning: New York, NY, USA, 2019.
  22. Lodish, H.F.; Berk, A.; Kaiser, C.; Krieger, M.; Scott, M.P.; Bretscher, A.; Ploegh, H.L.; Matsudaira, P.T. Molecular Cell Biology, 6th ed.; W.H. Freeman: New York, NY, USA, 2008.
  23. Jalloh, I.; Carpenter, K.L.H.; Grice, P.; Howe, D.J.; Mason, A.; Gallagher, C.N.; Helmy, A.; Murphy, M.P.; Menon, D.K.; Carpenter, T.A.; et al. Glycolysis and the Pentose Phosphate Pathway after Human Traumatic Brain Injury: Microdialysis Studies Using 1,2-13C2 Glucose. J. Cereb. Blood Flow Metab. 2015, 35, 111–120.
  24. Jalloh, I.; Helmy, A.; Howe, D.J.; Shannon, R.J.; Grice, P.; Mason, A.; Gallagher, C.N.; Murphy, M.P.; Pickard, J.D.; Menon, D.K.; et al. A Comparison of Oxidative Lactate Metabolism in Traumatically Injured Brain and Control Brain. J. Neurotrauma 2018, 35, 2025–2035.
  25. Hutchinson, P.J.; Jalloh, I.; Helmy, A.; Carpenter, K.L.; Rostami, E.; Bellander, B.M.; Boutelle, M.G.; Chen, J.W.; Claassen, J.; Dahyot-Fizelier, C.; et al. Consensus statement from the 2014 International Microdialysis Forum. Intensive Care Med. 2015, 41, 1517–1528.
  26. Gallagher, C.N.; Carpenter, K.L.; Grice, P.; Howe, D.J.; Mason, A.; Timofeev, I.; Menon, D.K.; Kirkpatrick, P.J.; Pickard, J.D.; Sutherland, G.R.; et al. The human brain utilizes lactate via the tricarboxylic acid cycle: A 13C-labelled microdialysis and high-resolution nuclear magnetic resonance study. Brain 2009, 132, 2839–2849.
  27. Guilfoyle, M.R.; Helmy, A.; Donnelly, J.; Stovell, M.G.; Timofeev, I.; Pickard, J.D.; Czosnyka, M.; Smielewski, P.; Menon, D.K.; Carpenter, K.L.H.; et al. Characterising the dynamics of cerebral metabolic dysfunction following traumatic brain injury: A microdialysis study in 619 patients. PLoS ONE 2021, 16, e0260291.
  28. Williamson, D.H.; Lund, P.; Krebs, H.A. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem. J. 1967, 103, 514–527.
  29. Vespa, P.; Boonyaputthikul, R.; McArthur, D.L.; Miller, C.; Etchepare, M.; Bergsneider, M.; Glenn, T.; Martin, N.; Hovda, D. Intensive insulin therapy reduces microdialysis glucose values without altering glucose utilization or improving the lactate/pyruvate ratio after traumatic brain injury. Crit. Care Med. 2006, 34, 850–856.
  30. Vespa, P.; McArthur, D.L.; Stein, N.; Huang, S.C.; Shao, W.; Filippou, M.; Etchepare, M.; Glenn, T.; Hovda, D.A. Tight glycemic control increases metabolic distress in traumatic brain injury: A randomized controlled within-subjects trial. Crit. Care Med. 2012, 40, 1923–1929.
  31. Oddo, M.; Schmidt, J.M.; Carrera, E.; Badjatia, N.; Connolly, E.S.; Presciutti, M.; Ostapkovich, N.D.; Levine, J.M.; Le Roux, P.; Mayer, S.A. Impact of tight glycemic control on cerebral glucose metabolism after severe brain injury: A microdialysis study. Crit. Care Med. 2008, 36, 3233–3238.
  32. Helbok, R.; Schmidt, J.M.; Kurtz, P.; Hanafy, K.A.; Fernandez, L.; Stuart, R.M.; Presciutti, M.; Ostapkovich, N.D.; Connolly, E.S.; Lee, K.; et al. Systemic glucose and brain energy metabolism after subarachnoid hemorrhage. Neurocrit. Care 2010, 12, 317–323.
  33. Rostami, E.; Bellander, B.M. Monitoring of glucose in brain, adipose tissue, and peripheral blood in patients with traumatic brain injury: A microdialysis study. J. Diabetes Sci. Technol. 2011, 5, 596–604.
  34. Tolias, C.M.; Reinert, M.; Seiler, R.; Gilman, C.; Scharf, A.; Bullock, M.R. Normobaric hyperoxia--induced improvement in cerebral metabolism and reduction in intracranial pressure in patients with severe head injury: A prospective historical cohort-matched study. J. Neurosurg. 2004, 101, 435–444.
  35. Nortje, J.; Coles, J.P.; Timofeev, I.; Fryer, T.D.; Aigbirhio, F.I.; Smielewski, P.; Outtrim, J.G.; Chatfield, D.A.; Pickard, J.D.; Hutchinson, P.J.; et al. Effect of hyperoxia on regional oxygenation and metabolism after severe traumatic brain injury: Preliminary findings. Crit. Care Med. 2008, 36, 273–281.
  36. Rockswold, S.B.; Rockswold, G.L.; Zaun, D.A.; Liu, J. A prospective, randomized Phase II clinical trial to evaluate the effect of combined hyperbaric and normobaric hyperoxia on cerebral metabolism, intracranial pressure, oxygen toxicity, and clinical outcome in severe traumatic brain injury. J. Neurosurg. 2013, 118, 1317–1328.
  37. Marion, D.W.; Puccio, A.; Wisniewski, S.R.; Kochanek, P.; Dixon, C.E.; Bullian, L.; Carlier, P. Effect of hyperventilation on extracellular concentrations of glutamate, lactate, pyruvate, and local cerebral blood flow in patients with severe traumatic brain injury. Crit. Care Med. 2002, 30, 2619–2625.
  38. Hutchinson, P.J.; Gupta, A.K.; Fryer, T.F.; Al-Rawi, P.G.; Chatfield, D.A.; Coles, J.P.; O’Connell, M.T.; Kett-White, R.; Minhas, P.S.; Aigbirhio, F.I.; et al. Correlation between Cerebral Blood Flow, Substrate Delivery, and Metabolism in Head Injury: A Combined Microdialysis and Triple Oxygen Positron Emission Tomography Study. J. Cereb. Blood Flow Metab. 2002, 22, 735–745.
  39. Sakowitz, O.W.; Stover, J.F.; Sarrafzadeh, A.S.; Unterberg, A.W.; Kiening, K.L. Effects of mannitol bolus administration on intracranial pressure, cerebral extracellular metabolites, and tissue oxygenation in severely head-injured patients. J. Trauma 2007, 62, 292–298.
  40. Chiu, C.; Xian, W.; Moss, C.F. Flying in silence: Echolocating bats cease vocalizing to avoid sonar jamming. Proc. Natl. Acad. Sci. USA 2008, 105, 13116–13121.
  41. Soukup, J.; Zauner, A.; Doppenberg, E.M.; Menzel, M.; Gilman, C.; Bullock, R.; Young, H.F. Relationship between brain temperature, brain chemistry and oxygen delivery after severe human head injury: The effect of mild hypothermia. Neurol. Res. 2002, 24, 161–168.
  42. Berger, C.; Schäbitz, W.R.; Georgiadis, D.; Steiner, T.; Aschoff, A.; Schwab, S. Effects of hypothermia on excitatory amino acids and metabolism in stroke patients: A microdialysis study. Stroke 2002, 33, 519–524.
  43. Ho, C.L.; Wang, C.M.; Lee, K.K.; Ng, I.; Ang, B.T. Cerebral oxygenation, vascular reactivity, and neurochemistry following decompressive craniectomy for severe traumatic brain injury. J. Neurosurg. 2008, 108, 943–949.
  44. Nagel, A.; Graetz, D.; Schink, T.; Frieler, K.; Sakowitz, O.; Vajkoczy, P.; Sarrafzadeh, A. Relevance of intracranial hypertension for cerebral metabolism in aneurysmal subarachnoid hemorrhage. Clinical article. J. Neurosurg. 2009, 111, 94–101.
  45. Papadimitriou, K.I.; Wang, C.; Rogers, M.L.; Gowers, S.A.N.; Leong, C.L.; Boutelle, M.G.; Drakakis, E.M. High-Performance Bioinstrumentation for Real-Time Neuroelectrochemical Traumatic Brain Injury Monitoring. Front. Hum. Neurosci. 2016, 10, 212.
  46. Pagkalos, I.; Rogers, M.; Boutelle, M.; Drakakis, E. A Higà Performance Application Specific Integrated Circuit for Electrical and Neurochemical Traumatic Brain Injury Monitoring. Chemphyschem 2018, 19, 1215–1225.
  47. Tageldeen, M.K.; Gowers, S.A.N.; Leong, C.L.; Boutelle, M.G.; Drakakis, E.M. Traumatic brain injury neuroelectrochemical monitoring: Behind-the-ear micro-instrument and cloud application. J. Neuroeng. Rehabil. 2020, 17, 114.
  48. Robbins, E.M.; Jaquins-Gerstl, A.; Fine, D.F.; Leong, C.L.; Dixon, C.E.; Wagner, A.K.; Boutelle, M.G.; Michael, A.C. Extended (10-Day) Real-Time Monitoring by Dexamethasone-Enhanced Microdialysis in the Injured Rat Cortex. ACS Chem. Neurosci. 2019, 10, 3521–3531.
  49. Alimagham, F.C.; Hutter, D.; Marco-García, N.; Gould, E.; Highland, V.H.; Huefner, A.; Giorgi-Coll, S.; Killen, M.J.; Zakrzewska, A.P.; Elliott, S.R.; et al. Cerebral Microdialysate Metabolite Monitoring using Mid-infrared Spectroscopy. Anal. Chem. 2021, 93, 11929–11936.
  50. Rogers, M.L.; Leong, C.L.; Gowers, S.A.; Samper, I.C.; Jewell, S.L.; Khan, A.; McCarthy, L.; Pahl, C.; Tolias, C.M.; Walsh, D.C.; et al. Simultaneous monitoring of potassium, glucose and lactate during spreading depolarization in the injured human brain–Proof of principle of a novel real-time neurochemical analysis system, continuous online microdialysis. J. Cereb. Blood Flow Metab. 2017, 37, 1883–1895.
  51. Gifford, E.K.; Robbins, E.M.; Jaquins-Gerstl, A.; Rerick, M.T.; Nwachuku, E.L.; Weber, S.G.; Boutelle, M.G.; Okonkwo, D.O.; Puccio, A.M.; Michael, A.C. Validation of Dexamethasone-Enhanced Continuous-Online Microdialysis for Monitoring Glucose for 10 Days after Brain Injury. ACS Chem. Neurosci. 2021, 12, 3588–3597.
  52. Hutchinson, P.J. Microdialysis in traumatic brain injury--methodology and pathophysiology. Acta Neurochir. Suppl. 2005, 95, 441–445.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 399
Revisions: 3 times (View History)
Update Date: 11 May 2022
1000/1000
Video Production Service