95 results on '"Maria Cristina Morganti-Kossmann"'
Search Results
2. Neuronal nuclear calcium signaling suppression of microglial reactivity is mediated by osteoprotegerin after traumatic brain injury
- Author
-
Albrecht Fröhlich, Florian Olde Heuvel, Rida Rehman, Sruthi Sankari Krishnamurthy, Shun Li, Zhenghui Li, David Bayer, Alison Conquest, Anna M. Hagenston, Albert Ludolph, Markus Huber-Lang, Tobias Boeckers, Bernd Knöll, Maria Cristina Morganti-Kossmann, Hilmar Bading, and Francesco Roselli
- Subjects
Traumatic brain injury ,Microglia ,Nuclear calcium ,Osteoprotegerin ,Synapses ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Traumatic brain injury (TBI) is characterized by massive changes in neuronal excitation, from acute excitotoxicity to chronic hyper- or hypoexcitability. Nuclear calcium signaling pathways are involved in translating changes in synaptic inputs and neuronal activity into discrete transcriptional programs which not only affect neuronal survival and synaptic integrity, but also the crosstalk between neurons and glial cells. Here, we report the effects of blunting neuronal nuclear calcium signals in the context of TBI. Methods We used AAV vectors to express the genetically encoded and nuclear-targeted calcium buffer parvalbumin (PV.NLS.mCherry) or the calcium/calmodulin buffer CaMBP4.mCherry in neurons only. Upon TBI, the extent of neuroinflammation, neuronal death and synaptic loss were assessed by immunohistochemistry and targeted transcriptome analysis. Modulation of the overall level of neuronal activity was achieved by PSAM/PSEM chemogenetics targeted to parvalbumin interneurons. The functional impact of neuronal nuclear calcium buffering in TBI was assessed by quantification of spontaneous whisking. Results Buffering neuronal nuclear calcium unexpectedly resulted in a massive and long-lasting increase in the recruitment of reactive microglia to the injury site, which was characterized by a disease-associated and phagocytic phenotype. This effect was accompanied by a substantial surge in synaptic loss and significantly reduced whisking activity. Transcriptome analysis revealed a complex effect of TBI in the context of neuronal nuclear calcium buffering, with upregulation of complement factors, chemokines and interferon-response genes, as well as the downregulation of synaptic genes and epigenetic regulators compared to control conditions. Notably, nuclear calcium buffering led to a substantial loss in neuronal osteoprotegerin (OPG), whereas stimulation of neuronal firing induced OPG expression. Viral re-expression of OPG resulted in decreased microglial recruitment and synaptic loss. OPG upregulation was also observed in the CSF of human TBI patients, underscoring its translational value. Conclusion Neuronal nuclear calcium signals regulate the degree of microglial recruitment and reactivity upon TBI via, among others, osteoprotegerin signals. Our findings support a model whereby neuronal activity altered after TBI exerts a powerful impact on the neuroinflammatory cascade, which in turn contributes to the overall loss of synapses and functional impairment.
- Published
- 2022
- Full Text
- View/download PDF
3. Monitoring the Neuroinflammatory Response Following Acute Brain Injury
- Author
-
Eric Peter Thelin, Tamara Tajsic, Frederick Adam Zeiler, David K. Menon, Peter J. A. Hutchinson, Keri L. H. Carpenter, Maria Cristina Morganti-Kossmann, and Adel Helmy
- Subjects
neuroinflammation ,traumatic brain injury ,subarachnoid hemorrhage ,multimodal monitoring ,secondary brain injury ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH) are major contributors to morbidity and mortality. Following the initial insult, patients may deteriorate due to secondary brain damage. The underlying molecular and cellular cascades incorporate components of the innate immune system. There are different approaches to assess and monitor cerebral inflammation in the neuro intensive care unit. The aim of this narrative review is to describe techniques to monitor inflammatory activity in patients with TBI and SAH in the acute setting. The analysis of pro- and anti-inflammatory cytokines in compartments of the central nervous system (CNS), including the cerebrospinal fluid and the extracellular fluid, represent the most common approaches to monitor surrogate markers of cerebral inflammatory activity. Each of these compartments has a distinct biology that reflects local processes and the cross-talk between systemic and CNS inflammation. Cytokines have been correlated to outcomes as well as ongoing, secondary injury progression. Alongside the dynamic, focal assay of humoral mediators, imaging, through positron emission tomography, can provide a global in vivo measurement of inflammatory cell activity, which reveals long-lasting processes following the initial injury. Compared to the innate immune system activated acutely after brain injury, the adaptive immune system is likely to play a greater role in the chronic phase as evidenced by T-cell-mediated autoreactivity toward brain-specific proteins. The most difficult aspect of assessing neuroinflammation is to determine whether the processes monitored are harmful or beneficial to the brain as accumulating data indicate a dual role for these inflammatory cascades following injury. In summary, the inflammatory component of the complex injury cascade following brain injury may be monitored using different modalities. Using a multimodal monitoring approach can potentially aid in the development of therapeutics targeting different aspects of the inflammatory cascade and improve the outcome following TBI and SAH.
- Published
- 2017
- Full Text
- View/download PDF
4. The scavenging chemokine receptor ACKR2 has a significant impact on acute mortality rate and early lesion development after traumatic brain injury.
- Author
-
Thomas M Woodcock, Tony Frugier, Tan Thanh Nguyen, Bridgette Deanne Semple, Nicole Bye, Matteo Massara, Benedetta Savino, Roberta Besio, Cristina Sobacchi, Massimo Locati, and Maria Cristina Morganti-Kossmann
- Subjects
Medicine ,Science - Abstract
The atypical chemokine receptor ACKR2 promotes resolution of acute inflammation by operating as a scavenger receptor for inflammatory CC chemokines in several experimental models of inflammatory disorders, however its role in the brain remains unclear. Based on our previous reports of increased expression of inflammatory chemokines and their corresponding receptors following traumatic brain injury (TBI), we hypothesised that ACKR2 modulates neuroinflammation following brain trauma and that its deletion exacerbates cellular inflammation and chemokine production. We demonstrate increased CCL2 and ACKR2 mRNA expression in post-mortem human brain, whereby ACKR2 mRNA levels correlated with later times post-TBI. This data is consistent with the transient upregulation of ACKR2 observed in mouse brain after closed head injury (CHI). As compared to WT animals, ACKR2-/- mice showed a higher mortality rate after CHI, while the neurological outcome in surviving mice was similar. At day 1 post-injury, ACKR2-/- mice displayed aggravated lesion volume and no differences in CCL2 expression and macrophage recruitment relative to WT mice. Reciprocal regulation of ACKR2 and CCL2 expression was explored in cultured astrocytes, which are recognized as the major source of CCL2 and also express ACKR2. ACKR2 mRNA increased as early as 2 hours after an inflammatory challenge in WT astrocytes. As expected, CCL2 expression also dramatically increased at 4 hours in WT astrocytes but was significantly lower in ACKR2-/- astrocytes, possibly indicating a co-regulation of CCL2 and ACKR2 in these cells. Conversely, in vivo, CCL2 mRNA/protein levels were increased similarly in ACKR2-/- and WT brains at 4 and 12 hours after CHI, in line with the lack of differences in cerebral macrophage recruitment and neurological recovery. In conclusion, ACKR2 is induced after TBI and has a significant impact on mortality and lesion development acutely following CHI, while its role in chemokine expression, macrophage activation, brain pathology, and neurological recovery at later time-points is minor. Concordant to evidence in multiple sclerosis experimental models, our data corroborate a distinct role for ACKR2 in cerebral inflammatory processes compared to its reported functions in peripheral tissues.
- Published
- 2017
- Full Text
- View/download PDF
5. Measurement of serum melatonin in intensive care unit patients: changes in traumatic brain injury, trauma and medical conditions
- Author
-
Marc A Seifman, Keith eGomes, Phuong eNguyen, Michael eBailey, Jeffrey V Rosenfeld, David J Cooper, and Maria Cristina Morganti-Kossmann
- Subjects
Melatonin ,circadian ,TBI ,Trauma ,Intensive Care Unit ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Melatonin is an endogenous hormone mainly produced by the pineal gland whose dysfunction leads to abnormal sleeping patterns. Changes in melatonin have been reported in acute traumatic brain injury (TBI), however the impact of environmental conditions typical of the intensive care unit (ICU) has not been assessed. The aim of this study was to compare daily melatonin production in three patient populations treated at the ICU to differentiate the role of TBI versus ICU conditions. Forty-five patients were recruited and divided into severe TBI, trauma without TBI, medical conditions without trauma and compared to healthy volunteers. Serum melatonin levels were measured at four daily intervals at 0400h, 1000h, 1600h and 2200h for 7 days post-ICU admission by commercial ELISA. The geometric mean concentrations (95% confidence intervals) of melatonin in these groups showed no difference being 8.3 (6.3-11.0), 9.3 (7.0-12.3) and 8.9 (6.6-11.9) pg/mL, respectively in TBI, trauma and intensive care cohorts. All of these patient groups demonstrated decreased melatonin concentrations when compared to control patients.This study suggests that TBI as well as ICU conditions, may have a role in the dysfunction of melatonin. Monitoring and possibly substituting melatonin acutely in these settings may assist in ameliorating longterm sleep dysfunction in all of these groups, and possibly contribute to reducing secondary brain injury in severe TBI.
- Published
- 2014
- Full Text
- View/download PDF
6. Guilty Molecules, Guilty Minds? The Conflicting Roles of the Innate Immune Response to Traumatic Brain Injury
- Author
-
Sarah Claire Hellewell and Maria Cristina Morganti-Kossmann
- Subjects
Pathology ,RB1-214 - Abstract
Traumatic brain injury (TBI) is a complex disease in the most complex organ of the body, whose victims endure lifelong debilitating physical, emotional, and psychosocial consequences. Despite advances in clinical care, there is no effective neuroprotective therapy for TBI, with almost every compound showing promise experimentally having disappointing results in the clinic. The complex and highly interrelated innate immune responses govern both the beneficial and deleterious molecular consequences of TBI and are present as an attractive therapeutic target. This paper discusses the positive, negative, and often conflicting roles of the innate immune response to TBI in both an experimental and clinical settings and highlights recent advances in the search for therapeutic candidates for the treatment of TBI.
- Published
- 2012
- Full Text
- View/download PDF
7. Met/HGFR triggers detrimental reactive microglia in TBI
- Author
-
Rida Rehman, Michael Miller, Sruthi Sankari Krishnamurthy, Jacob Kjell, Lobna Elsayed, Stefanie M. Hauck, Florian olde Heuvel, Alison Conquest, Akila Chandrasekar, Albert Ludolph, Tobias Boeckers, Medhanie A. Mulaw, Magdalena Goetz, Maria Cristina Morganti-Kossmann, Aya Takeoka, and Francesco Roselli
- Subjects
Neuroscience [CP] ,phosphorylation ,traumatic brain injury ,HGFR ,microglia ,antibody array ,General Biochemistry, Genetics and Molecular Biology ,neuroinflammation ,Mice, Inbred C57BL ,VEGFR ,Mice ,Disease Models, Animal ,Neuroscience ,Hgfr ,Met ,Microglia ,Neuroinflammation ,Phosphorylation ,Proteomics ,Traumatic Brain Injury ,Vegfr [Antibody Array ,Btk ,Cp] ,proteomics ,Brain Injuries, Traumatic ,Humans ,Animals ,ddc:610 ,Signal Transduction - Abstract
The complexity of signaling events and cellular responses unfolding in neuronal, glial, and immune cells upon traumatic brain injury (TBI) constitutes an obstacle in elucidating pathophysiological links and targets for intervention. We use array phosphoproteomics in a murine mild blunt TBI to reconstruct the temporal dynamics of tyrosine-kinase signaling in TBI and then scrutinize the large-scale effects of perturbation of Met/HGFR, VEGFR1, and Btk signaling by small molecules. We show Met/HGFR as a selective modifier of early microglial response and that Met/HGFR blockade prevents the induction of microglial inflammatory mediators, of reactive microglia morphology, and TBI-associated responses in neurons and vasculature. Both acute and prolonged Met/HGFR inhibition ameliorate neuronal survival and motor recovery. Early elevation of HGF itself in the cerebrospinal fluid of TBI patients suggests that this mechanism has translational value in human subjects. Our findings identify Met/HGFR as a modulator of early neuroinflammation in TBI with promising translational potential. ispartof: Cell Reports vol:41 issue:13 ispartof: location:United States status: published
- Published
- 2022
8. Interleukin-13 and its receptor are synaptic proteins involved in plasticity and neuroprotection
- Author
-
Shun Li, Florian olde Heuvel, Rida Rehman, Zhenghui Li, Oumayma Aousji, Albrecht Froehlich, Wanhong Zhang, Alison Conquest, Sarah Woelfle, Michael Schoen, Caitlin O’Meara, Richard Lee Reinhardt, David Voehringer, Jan Kassubek, Albert Ludolph, Markus Huber-Lang, Bernd Knöll, Maria Cristina Morganti-Kossmann, Tobias Boeckers, and Francesco Roselli
- Subjects
nervous system - Abstract
Immune system molecules are expressed by neurons, often for unknown functions. We have identified IL-13 and its receptor IL-13Ra1 as neuronal, synaptic proteins in mouse, rat, and human brains, whose engagement upregulates the phosphorylation of NMDAR and AMPAR subunits and, in turn, increases synaptic activity and CREB-mediated transcription. We demonstrate that increased IL-13 is a hallmark of traumatic brain injury (TBI) in mice as well as in two distinct cohorts of human patients. We also provide evidence that IL-13 upregulation protects neurons from excitotoxic death. We show IL-13 upregulation occurring in several cohorts of human brain samples and in CSF. Thus, IL-13 is a previously unrecognized physiological modulator of synaptic physiology of neuronal origin, with implications for the establishment of synaptic plasticity and the survival of neurons under injury conditions. Furthermore, we suggest that the neuroprotection afforded through the upregulation of IL-13 represents a new entry point for interventions in the pathophysiology of TBI.
- Published
- 2021
9. EPO treatment does not alter acute serum profiles of GFAP and S100B after TBI: A brief report on the Australian EPO-TBI clinical trial
- Author
-
Lorraine Little, Jasmin Board, Maria Cristina Morganti-Kossmann, Shirley Vallance, Rinaldo Bellomo, David James Cooper, Sarah C. Hellewell, and Alison Conquest
- Subjects
Adult ,Male ,medicine.medical_specialty ,Traumatic brain injury ,S100 Calcium Binding Protein beta Subunit ,Placebo ,Gastroenterology ,03 medical and health sciences ,Subcutaneous injection ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,Brain Injuries, Traumatic ,Glial Fibrillary Acidic Protein ,Medicine ,Humans ,Erythropoietin ,Glial fibrillary acidic protein ,biology ,business.industry ,Australia ,Epoetin alfa ,General Medicine ,Middle Aged ,medicine.disease ,Prognosis ,Clinical trial ,nervous system ,Neurology ,030220 oncology & carcinogenesis ,biology.protein ,Biomarker (medicine) ,Surgery ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Biomarkers ,medicine.drug - Abstract
Purpose To determine the diagnostic and prognostic value of glial fibrillary acidic protein (GFAP) and S100B after traumatic brain injury (TBI) in an Erythropoietin (EPO) clinical trial and examine whether EPO therapy reduces biomarker concentrations. Materials and Methods Forty-four patients with moderate-to-severe TBI were enrolled to a sub-study of the EPO-TBI trial. Patients were randomized to either Epoetin alfa 40,000 IU or 1 ml sodium chloride 0.9 as subcutaneous injection within 24 h of TBI. Results GFAP and S100B were measured in serum by ELISA from D0 (within 24 h of injury, prior to EPO/vehicle administration) to D5. Biomarker concentrations were compared between injury severities, diffuse vs. focal TBI, 6-month outcome scores (GOS-E) and EPO or placebo treatments. At D0 GFAP was significantly higher than S100B (951 pg/mL vs. 476 pg/mL, p = 0.018). ROC analysis of S100B at 1D post-injury distinguished favorable vs. unfavorable outcomes (area under the curve = 0.73; p = 0.01). EPO did not reduce concentration of either biomarker. Conclusions Elevated serum concentrations of GFAP and S100B after TBI reflect a robust, acute glial response to injury. Consistent with lack of improved outcome in TBI patients treated with EPO and prior findings on neuronal and axonal markers, glial biomarker concentrations and acute profiles were not affected by EPO.
- Published
- 2020
10. Erythropoietin Does Not Alter Serum Profiles of Neuronal and Axonal Biomarkers After Traumatic Brain Injury
- Author
-
Jasmine Board, Lorraine Little, Nicole Bye, Maria Cristina Morganti-Kossmann, Irina Madorsky, Stefania Mondello, David James Cooper, Firas Kobeissy, Gerry Shaw, Shirley Vallance, Rinaldo Bellomo, Alison Conquest, Jay V. Deng, and Sarah C. Hellewell
- Subjects
Adult ,Male ,0301 basic medicine ,Neurofilament ,Traumatic brain injury ,Enzyme-Linked Immunosorbent Assay ,Pharmacology ,Critical Care and Intensive Care Medicine ,Biomarker, Erythropoietin, Phosphorylated neurofilament heavy-chain, Traumatic brain injury, Ubiquitin carboxy-terminal hydrolase L1 ,03 medical and health sciences ,0302 clinical medicine ,Double-Blind Method ,Ubiquitin ,Neurofilament Proteins ,Brain Injuries, Traumatic ,Humans ,Medicine ,Glasgow Coma Scale ,Prospective Studies ,Erythropoietin ,biology ,business.industry ,Australia ,Epoetin alfa ,Middle Aged ,medicine.disease ,Ubiquitin carboxy-terminal hydrolase L1 ,Epoetin Alfa ,Clinical trial ,030104 developmental biology ,biology.protein ,Biomarker (medicine) ,Female ,business ,Ubiquitin Thiolesterase ,Biomarkers ,030217 neurology & neurosurgery ,medicine.drug - Abstract
To determine profiles of serum ubiquitin carboxy-terminal hydrolase L1 and phosphorylated neurofilament heavy-chain, examine whether erythropoietin administration reduce their concentrations, and whether biomarkers discriminate between erythropoietin and placebo treatment groups.Single-center, prospective observational study.A sub-study of the erythropoietin-traumatic brain injury clinical trial, conducted at the Alfred Hospital, Melbourne, Australia.Forty-four patients with moderate-to-severe traumatic brain injury.Epoetin alfa 40,000 IU or 1 mL sodium chloride 0.9 as subcutaneous injection within 24 hours of traumatic brain injury.Ubiquitin carboxy-terminal hydrolase L1, phosphorylated neurofilament heavy-chain, and erythropoietin concentrations were measured in serum by enzyme-linked immunosorbent assay from D0 (within 24 hr of injury, prior to erythropoietin/vehicle administration) to D5. Biomarker concentrations were compared between injury severities, diffuse versus focal traumatic brain injury and erythropoietin or placebo treatment groups. Ubiquitin carboxy-terminal hydrolase L1 peaked at 146.0 ng/mL on D0, significantly decreased to 84.30 ng/mL on D1, and declined thereafter. Phosphorylated neurofilament heavy-chain levels were lowest at D0 and peaked on D5 at 157.9 ng/mL. D0 ubiquitin carboxy-terminal hydrolase L1 concentrations were higher in diffuse traumatic brain injury. Peak phosphorylated neurofilament heavy-chain levels on D3 and D4 correlated with Glasgow Outcome Score-Extended, predicting poor outcome. Erythropoietin did not reduce concentrations of ubiquitin carboxy-terminal hydrolase L1 or phosphorylated neurofilament heavy-chain.Serum ubiquitin carboxy-terminal hydrolase L1 and phosphorylated neurofilament heavy-chain increase after traumatic brain injury reflecting early neuronal and progressive axonal injury. Consistent with lack of improved outcome in traumatic brain injury patients treated with erythropoietin, biomarker concentrations and profiles were not affected by erythropoietin. Pharmacokinetics of erythropoietin suggest that the dose given was possibly too low to exert neuroprotection.
- Published
- 2018
11. Targeted therapeutic mild hypercapnia after cardiac arrest: A phase II multi-centre randomised controlled trial (the CCC trial)
- Author
-
Suzanne Eliott, Johan Mårtensson, Leah Peck, Nerina Harley, Alice Pébay, Stephen J Warrillow, Satoshi Suzuki, Tammy Lamac, Rachael Parke, Aiko Tanaka, Pauline Galt, John S. Archer, Stephen Bernard, Maria Cristina Morganti-Kossmann, Carol L. Hodgson, Alison Conquest, Eileen Gilder, Gopal Taori, Rinaldo Bellomo, Michael Bailey, Lianne McCarthy, Glenn M Eastwood, Antoine G. Schneider, Dion Stub, Deborah Barge, Shay McGuinness, Helen Young, and Graeme K Hart
- Subjects
Male ,medicine.medical_specialty ,medicine.medical_treatment ,S100 Calcium Binding Protein beta Subunit ,030204 cardiovascular system & hematology ,Emergency Nursing ,law.invention ,Hypercapnia ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Intensive care ,medicine ,Humans ,Glasgow Coma Scale ,Cardiopulmonary resuscitation ,Normocapnia ,Mechanical ventilation ,Analysis of Variance ,business.industry ,030208 emergency & critical care medicine ,Length of Stay ,Middle Aged ,Respiration, Artificial ,Heart Arrest ,Surgery ,Intensive Care Units ,Cerebral blood flow ,Phosphopyruvate Hydratase ,Anesthesia ,Emergency Medicine ,Female ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers - Abstract
Background In intensive care observational studies, hypercapnia after cardiac arrest (CA) is independently associated with improved neurological outcome. However, the safety and feasibility of delivering targeted therapeutic mild hypercapnia (TTMH) for such patients is untested. Methods In a phase II safety and feasibility multi-centre, randomised controlled trial, we allocated ICU patients after CA to 24h of targeted normocapnia (TN) (PaCO 2 35–45mmHg) or TTMH (PaCO 2 50–55mmHg). The primary outcome was serum neuron specific enolase (NSE) and S100b protein concentrations over the first 72h assessed in the first 50 patients surviving to day three. Secondary end-points included global measure of function assessment at six months and mortality for all patients. Results We enrolled 86 patients. Their median age was 61 years (58, 64 years) and 66 (79%) were male. Of these, 50 patients (58%) survived to day three for full biomarker assessment. NSE concentrations increased in the TTMH group ( p =0.02) and TN group ( p =0.005) over time, with the increase being significantly more pronounced in the TN group ( p (interaction)=0.04). S100b concentrations decreased over time in the TTMH group ( p p =0.68). However, the S100b change over time did not differ between the groups ( p (interaction)=0.23). At six months, 23 (59%) TTMH patients had good functional recovery compared with 18 (46%) TN patients. Hospital mortality occurred in 11 (26%) TTMH patients and 15 (37%) TN patients ( p =0.31). Conclusions In CA patients admitted to the ICU, TTMH was feasible, appeared safe and attenuated the release of NSE compared with TN. These findings justify further investigation of this novel treatment.
- Published
- 2016
12. Therapies negating neuroinflammation after brain trauma
- Author
-
Maria Cristina Morganti-Kossmann, Sarah C. Hellewell, and Bridgette D. Semple
- Subjects
0301 basic medicine ,Neuroimmunomodulation ,Traumatic brain injury ,Anti-Inflammatory Agents ,Inflammation ,Therapeutic targeting ,Neuroprotection ,03 medical and health sciences ,0302 clinical medicine ,Brain Injuries, Traumatic ,medicine ,Animals ,Humans ,Molecular Biology ,Brain trauma ,Neuroinflammation ,Clinical Trials as Topic ,business.industry ,General Neuroscience ,medicine.disease ,Clinical trial ,Neuroprotective Agents ,030104 developmental biology ,Clinical research ,Neurology (clinical) ,medicine.symptom ,business ,Neuroscience ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Traumatic brain injury (TBI) elicits a complex secondary injury response, with neuroinflammation as a crucial central component. Long thought to be solely a deleterious factor, the neuroinflammatory response has recently been shown to be far more intricate, with both beneficial and detrimental consequences depending on the timing, magnitude and specific immune composition of the response post-injury. Despite extensive preclinical and clinical research into mechanisms of secondary injury after TBI, no effective neuroprotective therapy has been identified, with potential candidates repeatedly proving disappointing in the clinic. The neuroinflammatory response offers a promising avenue for therapeutic targeting, aiming to quell the deleterious consequences without influencing its function in providing a neurotrophic environment supportive of repair. The present review firstly describes the findings of recent clinical trials that aimed to modulate inflammation as a means of neuroprotection. Secondly, we discuss promising multifunctional and single-target anti-inflammatory candidates either currently in trial, or with ample experimental evidence supporting clinical application. This article is part of a Special Issue entitled SI:Brain injury and recovery.
- Published
- 2016
13. Environmental Enrichment Attenuates Traumatic Brain Injury: Induced Neuronal Hyperexcitability in Supragranular Layers of Sensory Cortex
- Author
-
Ramesh Rajan, Maria Cristina Morganti-Kossmann, Simone F. Carron, Sarah C. Hellewell, Victoria P.A. Johnstone, Dasuni S. Alwis, and Edwin BingBing Yan
- Subjects
0301 basic medicine ,Male ,Population ,Sensory system ,Environment ,Somatosensory system ,Rats, Sprague-Dawley ,03 medical and health sciences ,Random Allocation ,0302 clinical medicine ,Neuroplasticity ,Brain Injuries, Traumatic ,medicine ,Premovement neuronal activity ,Animals ,Sensory cortex ,education ,Neurons ,Environmental enrichment ,education.field_of_study ,Neuronal Plasticity ,Behavior, Animal ,business.industry ,Somatosensory Cortex ,Barrel cortex ,Rats ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Vibrissae ,Cortical Excitability ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
We have previously demonstrated that traumatic brain injury (TBI) induces significant long-term neuronal hyperexcitability in supragranular layers of sensory cortex, coupled with persistent sensory deficits. Hence, we aimed to investigate whether brain plasticity induced by environmental enrichment (EE) could attenuate abnormal neuronal and sensory function post-TBI. TBI (n = 22) and sham control (n = 21) animals were randomly assigned housing in either single or enriched conditions for 7-9 weeks. Then, in terminal experiments, extracellular recordings were obtained from barrel cortex neurons in response to whisker motion, including those mimicking motion in awake animals undertaking different tasks. Long-term EE exposure (6 weeks) attenuated TBI-induced hyperexcitability in layers 2-3, such that neuronal activity in TBI animals exposed to EE was restored to control levels. Little to no EE-induced changes in population neuronal responses occurred in input layer 4 and output layer 5. However, single-cell responses demonstrated EE-induced hypoexcitation in L4 post-TBI. EE was also able to fully ameliorate sensory hypersensitivity post-TBI, although it was not found to improve motor function. Long-term enrichment post-TBI induces changes at both the population and single-cell level in the sensory cortex, where EE may act to restore the excitation/inhibition balance in supragranular cortical layers.
- Published
- 2015
14. Attenuated neurological deficit, cell death and lesion volume in Fas-mutant mice is associated with altered neuroinflammation following traumatic brain injury
- Author
-
Jenna M. Ziebell, Thomas Kossmann, Bridgette D. Semple, Nicole Bye, and Maria Cristina Morganti-Kossmann
- Subjects
Mice, Inbred MRL lpr ,Fas Ligand Protein ,Time Factors ,Traumatic brain injury ,medicine.medical_treatment ,Inflammation ,Brain damage ,Biology ,Neuroprotection ,Fas ligand ,Mice ,Glial Fibrillary Acidic Protein ,In Situ Nick-End Labeling ,medicine ,Animals ,Humans ,fas Receptor ,Molecular Biology ,Neuroinflammation ,Analysis of Variance ,Cell Death ,Microglia ,Caspase 3 ,Tumor Necrosis Factor-alpha ,Macrophages ,General Neuroscience ,medicine.disease ,Disease Models, Animal ,Cytokine ,medicine.anatomical_structure ,Brain Injuries ,Immunoglobulin G ,Phosphopyruvate Hydratase ,Mutation ,Immunology ,Cytokines ,Neurology (clinical) ,Nervous System Diseases ,medicine.symptom ,Developmental Biology - Abstract
Progressive neurodegeneration following traumatic brain injury (TBI) involves the Fas and TNF-receptor1 protein systems which have been implicated in mediating delayed cell death. In this study, we used two approaches to assess whether inhibition of these pathways reduced secondary brain damage and neurological deficits after TBI. Firstly, we investigated whether the expression of non-functional Fas in lpr mice subjected to TBI altered tissue damage and neurological outcome. Compared to wild-type, lpr mice showed improved neurological deficit (p=0.0009), decreased lesion volume (p=0.017), number of TUNEL+ cells (p=0.011) and caspase-3+ cells (p=0.007). Changes in cellular inflammation and cytokine production were also compared between mouse strains. Accumulation of macrophages/microglia occurred earlier in lpr mice, likely due to enhanced production of the chemotactic mediators IL-12(p40) and MCP-1 (p
- Published
- 2011
15. Animal models of traumatic brain injury: Is there an optimal model to reproduce human brain injury in the laboratory?
- Author
-
Maria Cristina Morganti-Kossmann, Nicole Bye, and Edwin BingBing Yan
- Subjects
Pathology ,medicine.medical_specialty ,Swine ,Traumatic brain injury ,Cost-Benefit Analysis ,Ischemia ,Poison control ,Brain damage ,Dogs ,Animal model ,Injury prevention ,medicine ,Animals ,Humans ,General Environmental Science ,business.industry ,Experimental model ,Australia ,Brain ,Reproducibility of Results ,Human brain ,medicine.disease ,Rats ,Disease Models, Animal ,medicine.anatomical_structure ,Brain Injuries ,Cats ,General Earth and Planetary Sciences ,medicine.symptom ,business ,Neuroscience - Abstract
Compared to other neurological diseases, the research surrounding traumatic brain injury (TBI) has a more recent history. The establishment and use of animal models of TBI remains vital to understand the pathophysiology of this highly complex disease. Such models share the ultimate goals of reproducing patterns of tissue damage observed in humans (thus rendering them clinically relevant), reproducible and highly standardised to allow for the manipulation of individual variables, and to finally explore novel therapeutics for clinical translation. There is no doubt that the similarity of cellular and molecular events observed in human and rodent TBI has reinforced the use of small animals for research. When confronted with the choice of the experimental model it becomes clear that the ideal animal model does not exist. This limitation derives from the fact that most models mimic either focal or diffuse brain injury, whereas the clinical reality suggests that each patient has an individual form of TBI characterised by various combinations of focal and diffuse patterns of tissue damage. This is additionally complicated by the occurrence of secondary insults such as hypotension, hypoxia, ischaemia, extracranial injuries, modalities of traumatic events, age, gender and heterogeneity of medical treatments and pre-existing conditions. This brief review will describe the variety of TBI models available for laboratory research beginning from the most widely used rodent models of focal brain trauma, to complex large species such as the pig. In addition, the models mimicking diffuse brain damage will be discussed in relation to the early primate studies until the use of most common rodent models to elucidate the intriguing and less understood pathology of axonal dysfunction. The most recent establishment of in vitro paradigms has complemented the in vivo modelling studies offering a further cellular and molecular insight of this pathology.
- Published
- 2010
16. Involvement of pro- and anti-inflammatory cytokines and chemokines in the pathophysiology of traumatic brain injury
- Author
-
Maria Cristina Morganti-Kossmann and Jenna M. Ziebell
- Subjects
Chemokine ,medicine.medical_specialty ,Neurology ,Traumatic brain injury ,Models, Neurological ,Anti-Inflammatory Agents ,Inflammation ,Review Article ,Neuroprotection ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Neuroinflammation ,Pharmacology ,biology ,business.industry ,Brain ,medicine.disease ,Pathophysiology ,Brain Injuries ,Immunology ,biology.protein ,Cytokines ,Neurology (clinical) ,Chemokines ,medicine.symptom ,business ,Homeostasis - Abstract
Despite dramatic improvements in the management of traumatic brain injury (TBI), to date there is no effective treatment available to patients, and morbidity and mortality remain high. The damage to the brain occurs in two phases, the initial primary phase being the injury itself, which is irreversible and amenable only to preventive measures to minimize the extent of damage, followed by an ongoing secondary phase, which begins at the time of injury and continues in the ensuing days to weeks. This delayed phase leads to a variety of physiological, cellular, and molecular responses aimed at restoring the homeostasis of the damaged tissue, which, if not controlled, will lead to secondary insults. The development of secondary brain injury represents a window of opportunity in which pharmaceutical compounds with neuroprotective properties could be administered. To establish effective treatments for TBI victims, it is imperative that the complex molecular cascades contributing to secondary injury be fully elucidated. One pathway known to be activated in response to TBI is cellular and humoral inflammation. Neuroinflammation within the injured brain has long been considered to intensify the damage sustained following TBI. However, the accumulated findings from years of clinical and experimental research support the notion that the action of inflammation may differ in the acute and delayed phase after TBI, and that maintaining limited inflammation is essential for repair. This review addresses the role of several cytokines and chemokines following focal and diffuse TBI, as well as the controversies around the use of therapeutic anti-inflammatory treatments versus genetic deletion of cytokine expression.
- Published
- 2010
17. Activation of the kynurenine pathway and increased production of the excitotoxin quinolinic acid following traumatic brain injury in humans
- Author
-
May Tan, Chai K. Lim, Tony Frugier, Gayathri Sundaram, David W. Walker, Gilles J. Guillemin, Edwin BingBing Yan, Maria Cristina Morganti-Kossmann, Benjamin Heng, and Jeffrey V. Rosenfeld
- Subjects
Male ,Kynurenine pathway ,Excitotoxicity ,Glasgow Outcome Scale ,medicine.disease_cause ,chemistry.chemical_compound ,0302 clinical medicine ,Kynurenic acid ,Cerebrospinal fluid ,Traumatic brain injury ,Quinolinic acid ,Kynurenine ,0303 health sciences ,General Neuroscience ,Tryptophan ,Brain ,Middle Aged ,Prognosis ,Neurology ,Female ,medicine.symptom ,Signal Transduction ,Adult ,medicine.medical_specialty ,Patients ,Adolescent ,Immunology ,Neurotoxins ,Brain damage ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,Internal medicine ,medicine ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,RNA, Messenger ,Tryptophan metabolism ,030304 developmental biology ,Aged ,Research ,Neurotoxicity ,medicine.disease ,Endocrinology ,chemistry ,Brain Injuries ,Case-Control Studies ,030217 neurology & neurosurgery ,Biomarkers - Abstract
During inflammation, the kynurenine pathway (KP) metabolises the essential amino acid tryptophan (TRP) potentially contributing to excitotoxicity via the release of quinolinic acid (QUIN) and 3-hydroxykynurenine (3HK). Despite the importance of excitotoxicity in the development of secondary brain damage, investigations on the KP in TBI are scarce. In this study, we comprehensively characterised changes in KP activation by measuring numerous metabolites in cerebrospinal fluid (CSF) from TBI patients and assessing the expression of key KP enzymes in brain tissue from TBI victims. Acute QUIN levels were further correlated with outcome scores to explore its prognostic value in TBI recovery. Methods Twenty-eight patients with severe TBI (GCS ≤ 8, three patients had initial GCS = 9–10, but rapidly deteriorated to ≤8) were recruited. CSF was collected from admission to day 5 post-injury. TRP, kynurenine (KYN), kynurenic acid (KYNA), QUIN, anthranilic acid (AA) and 3-hydroxyanthranilic acid (3HAA) were measured in CSF. The Glasgow Outcome Scale Extended (GOSE) score was assessed at 6 months post-TBI. Post-mortem brains were obtained from the Australian Neurotrauma Tissue and Fluid Bank and used in qPCR for quantitating expression of KP enzymes (indoleamine 2,3-dioxygenase-1 (IDO1), kynurenase (KYNase), kynurenine amino transferase-II (KAT-II), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3HAO) and quinolinic acid phosphoribosyl transferase (QPRTase) and IDO1 immunohistochemistry. Results In CSF, KYN, KYNA and QUIN were elevated whereas TRP, AA and 3HAA remained unchanged. The ratios of QUIN:KYN, QUIN:KYNA, KYNA:KYN and 3HAA:AA revealed that QUIN levels were significantly higher than KYN and KYNA, supporting increased neurotoxicity. Amplified IDO1 and KYNase mRNA expression was demonstrated on post-mortem brains, and enhanced IDO1 protein coincided with overt tissue damage. QUIN levels in CSF were significantly higher in patients with unfavourable outcome and inversely correlated with GOSE scores. Conclusion TBI induced a striking activation of the KP pathway with sustained increase of QUIN. The exceeding production of QUIN together with increased IDO1 activation and mRNA expression in brain-injured areas suggests that TBI selectively induces a robust stimulation of the neurotoxic branch of the KP pathway. QUIN’s detrimental roles are supported by its association to adverse outcome potentially becoming an early prognostic factor post-TBI.
- Published
- 2015
18. Low T3 Syndrome in Head-Injured Patients is Associated with Prolonged Suppression of Markers of Cell-Mediated Immune Response
- Author
-
Maria Cristina Morganti-Kossmann, Helen I. Joller-Jemelka, Thomas Kossmann, Philipp M. Lenzlinger, and Volkmar Hans
- Subjects
medicine.medical_specialty ,Triiodothyronine ,business.industry ,Traumatic brain injury ,Glasgow Coma Scale ,Neopterin ,Low T3 Syndrome ,medicine.disease ,chemistry.chemical_compound ,Endocrinology ,Cerebrospinal fluid ,Immune system ,chemistry ,Internal medicine ,medicine ,Surgery ,business ,Euthyroid sick syndrome - Abstract
To clarify the association between disturbed thyroid hormone metabolism (low T3 syndrome) and release of cytokines and markers of cell-mediated immune response. Concentrations of cytokines as well as of thyroid hormones were determined in 32 patients suffering from severe traumatic brain injury: interleukin-( IL-)1, IL-6, IL-10, tumor necrosis factor, transforming growth factor-(TGF-)β, soluble interleukin-2 receptor (sIL-2R), neopterin, and β2-microglobulin (β2m) in serum and cerebrospinal fluid; triiodothyronine (T3), free T3, thyroxine (T4), free T4, thyrotropin, thyroxine-binding globulin, and albumin in serum. Additionally, clinical parameters were assessed: Glasgow Coma Score, CT scan, intracranial pressure, Glasgow Outcome Score, and occurrence of pneumonia. Among 31 patients with a low T3 syndrome, those with additional low serum T4 levels (n = 13) showed a prolonged suppression of serum β2m, neopterin, and sIL-2R, and a higher secondary increase of serum β2m, neopterin, and TGF-β, as well as lower T3 levels (all p < 0.05). These patients also had a longer stay in the intensive care unit (34 ± 6 days vs. 22 ± 12 days; p = 0.008). Increased levels of β2m correlated with a preceding decrease of thyrotropin (cerebrospinal fluid: r = –0.53; p = 0.004; serum: r = –0.41; p = 0.029). Associations of thyroid hormone metabolism with either other cytokines or with clinical parameters were not detected. These results show that low T3 syndrome is a very common pathophysiological feature after severe traumatic brain injury. The association of a low T3 syndrome in combination with low serum T4 levels, with an altered time course of markers of cell-mediated immunity led the authors to hypothesize that a disturbed thyroid hormone metabolism may be interrelated with a prolonged cellular immune dysfunction after traumatic brain injury.
- Published
- 2005
19. Elevated Intracranial IL-18 in Humans and Mice after Traumatic Brain Injury and Evidence of Neuroprotective Effects of IL-18—Binding Protein after Experimental Closed Head Injury
- Author
-
Otmar Trentz, Mario Rancan, Charles A. Dinarello, Thomas Kossmann, Claudio A. Redaelli, Ido Yatsiv, Daniel Perez, Daniela Novick, Philip F. Stahel, Viviane I. Otto, Esther Shohami, Menachem Rubinstein, and Maria Cristina Morganti-Kossmann
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pathology ,Traumatic brain injury ,Inflammation ,Neuroprotection ,030218 nuclear medicine & medical imaging ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Head Injuries, Closed ,Internal medicine ,medicine ,Animals ,Humans ,Neuroinflammation ,Glycoproteins ,business.industry ,Interleukin-18 ,Brain ,Interleukin ,Middle Aged ,medicine.disease ,Recombinant Proteins ,Mice, Inbred C57BL ,Neuroprotective Agents ,Endocrinology ,Neurology ,Brain Injuries ,Closed head injury ,Intercellular Signaling Peptides and Proteins ,Female ,Interleukin 18 ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery - Abstract
Proinflammatory cytokines are important mediators of neuroinflammation after traumatic brain injury. The role of interleukin (IL)-18, a new member of the IL-1 family, in brain trauma has not been reported to date. The authors investigated the posttraumatic release of IL-18 in murine brains following experimental closed head injury (CHI) and in CSF of CHI patients. In the mouse model, intracerebral IL-18 was induced within 24 hours by ether anesthesia and sham operation. Significantly elevated levels of IL-18 were detected at 7 days after CHI and in human CSF up to 10 days after trauma. Published data imply that IL-18 may play a pathophysiological role in inflammatory CNS diseases; therefore its inhibition may ameliorate outcome after CHI. To evaluate the functional aspects of IL-18 in the injured brain, mice were injected systemically with IL-18–binding protein (IL-18BP), a specific inhibitor of IL-18, 1 hour after trauma. IL-18BP—treated mice showed a significantly improved neurological recovery by 7 days, accompanied by attenuated intracerebral IL-18 levels. This demonstrates that inhibition of IL-18 is associated with improved recovery. However, brain edema at 24 hours was not influenced by IL-18BP, suggesting that inflammatory mediators other than IL-18 induce the early detrimental effects of intracerebral inflammation.
- Published
- 2002
20. Inflammatory response in acute traumatic brain injury: a double-edged sword
- Author
-
Mario Rancan, Maria Cristina Morganti-Kossmann, Philip F. Stahel, and Thomas Kossmann
- Subjects
Traumatic brain injury ,Central nervous system ,Inflammation ,Brain damage ,Critical Care and Intensive Care Medicine ,Mice ,Transforming Growth Factor beta ,Leukocytes ,medicine ,Animals ,Humans ,Neuroinflammation ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,business.industry ,Regeneration (biology) ,Intercellular Adhesion Molecule-1 ,medicine.disease ,Interleukin-10 ,Cellular infiltration ,medicine.anatomical_structure ,Brain Injuries ,Immunology ,Cytokines ,Encephalitis ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Neuroscience - Abstract
Inflammation is an important part of the pathophysiology of traumatic brain injury. Although the central nervous system differs from the other organs because of the almost complete isolation from the blood stream mediated by the blood-brain barrier, the main steps characterizing the immune activation within the brain follow a scenario similar to that in other organs. The key players in these processes are the numerous immune mediators released within minutes of the primary injury. They guide a sequence of events including expression of adhesion molecules, cellular infiltration, and additional secretion of inflammatory molecules and growth factors, resulting in either regeneration or cell death. The question is this: to what extent is inflammation beneficial for the injured brain tissue, and how does it contribute to secondary brain damage and progressive neuronal loss? This review briefly reports recent evidence supporting the dual, the beneficial, or the deleterious role of neuroinflammation after traumatic brain injury.
- Published
- 2002
21. The production of macrophage inflammatory protein-2 induced by soluble intercellular adhesion molecule-1 in mouse astrocytes is mediated by src tyrosine kinases and p42/44 mitogen-activated protein kinase
- Author
-
Vivianne I Otto, Urs Gilli, Stefan Frentzel, Gerd Folkers, Sergio M. Gloor, Andreas E Hein, Otmar Trentz, Thomas Kossmann, Maria Cristina Morganti-Kossmann, and Emerita Ammann
- Subjects
MAPK/ERK pathway ,Neutrophils ,p38 mitogen-activated protein kinases ,Chemokine CXCL2 ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,Receptor tyrosine kinase ,Mice ,Cellular and Molecular Neuroscience ,Animals ,Humans ,Protein Isoforms ,Phosphorylation ,Cells, Cultured ,Mice, Knockout ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,biology ,Tumor Necrosis Factor-alpha ,Chemotaxis ,Monokines ,Intercellular Adhesion Molecule-1 ,Cell biology ,Enzyme Activation ,Mice, Inbred C57BL ,src-Family Kinases ,Astrocytes ,Culture Media, Conditioned ,Mitogen-activated protein kinase ,biology.protein ,Mitogen-Activated Protein Kinases ,Signal transduction ,Tyrosine kinase ,Protein Binding ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Severe traumatic brain injury stimulates the release of soluble intercellular adhesion molecule-1 (sICAM-1) into CSF. Studies in cultured mouse astrocytes suggest that sICAM-1 induces the production of macrophage inflammatory protein-2 (MIP-2). In the present study, we investigated the underlying mechanisms for MIP-2 induction. sICAM-1 induced MIP-2 in astrocytes lacking membrane-bound ICAM-1, indicating that its action is due to heterophilic binding to an undescribed receptor rather than homophilic binding to surface ICAM-1. Signal transduction may be mediated by src tyrosine kinases, as the src tyrosine kinase inhibitors herbimycin A and PP2 abolished MIP-2 induction by sICAM-1. Phosphorylation of p42/44 mitogen-activated protein kinase (MAPK), but not of p38 MAPK, occurred further downstream, as evidenced by western blot analysis combined with the use of herbimycin A and specific MAPK inhibitors. By contrast, induction of MIP-2 by tumour necrosis factor-alpha (TNF-alpha) involved both p42/44 MAPK and p38 MAPK. Following stimulation with either sICAM-1 or TNF-alpha, astrocyte supernatants promoted chemotaxis of human neutrophils and incubation of these supernatants with anti-MIP-2 antibodies more efficiently suppressed the migration induced by sICAM-1 than by TNF-alpha. These results show that sICAM-1 induces the production of biologically active MIP-2 in astrocytes by heterophilic binding to an undefined receptor and activation of src tyrosine kinases and p42/44 MAPK.
- Published
- 2002
22. The scavenging chemokine receptor ACKR2 has a significant impact on acute mortality rate and early lesion development after traumatic brain injury
- Author
-
Roberta Besio, Matteo Massara, Cristina Sobacchi, Tony Frugier, Bridgette D. Semple, Nicole Bye, Benedetta Savino, Tan Thanh Nguyen, Massimo Locati, Thomas M. Woodcock, and Maria Cristina Morganti-Kossmann
- Subjects
Male ,0301 basic medicine ,Macroglial Cells ,Chemokine ,Pathology ,Critical Care and Emergency Medicine ,Traumatic Brain Injury ,lcsh:Medicine ,Pathology and Laboratory Medicine ,White Blood Cells ,Mice ,Chemokine receptor ,0302 clinical medicine ,Animal Cells ,Brain Injuries, Traumatic ,Medicine and Health Sciences ,Brain Damage ,lcsh:Science ,Immune Response ,Musculoskeletal System ,Trauma Medicine ,Cells, Cultured ,Chemokine CCL2 ,Mammals ,Multidisciplinary ,biology ,Chemotaxis ,Eukaryota ,Brain ,Human brain ,Up-Regulation ,Cell Motility ,medicine.anatomical_structure ,Neurology ,Vertebrates ,Receptors, Chemokine ,Cellular Types ,Chemokines ,Anatomy ,medicine.symptom ,Traumatic Injury ,Research Article ,medicine.medical_specialty ,Traumatic brain injury ,Immune Cells ,Immunology ,Glial Cells ,Inflammation ,Rodents ,Bone and Bones ,Lesion ,03 medical and health sciences ,Signs and Symptoms ,Diagnostic Medicine ,Internal medicine ,medicine ,Animals ,Humans ,RNA, Messenger ,Mortality ,Skeleton ,Neuroinflammation ,Blood Cells ,business.industry ,Macrophages ,Skull ,lcsh:R ,Organisms ,Biology and Life Sciences ,Cell Biology ,Recovery of Function ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Astrocytes ,Amniotes ,Closed head injury ,biology.protein ,lcsh:Q ,business ,Neurotrauma ,Gene Deletion ,030217 neurology & neurosurgery - Abstract
The atypical chemokine receptor ACKR2 promotes resolution of acute inflammation by operating as a scavenger receptor for inflammatory CC chemokines in several experimental models of inflammatory disorders, however its role in the brain remains unclear. Based on our previous reports of increased expression of inflammatory chemokines and their corresponding receptors following traumatic brain injury (TBI), we hypothesised that ACKR2 modulates neuroinflammation following brain trauma and that its deletion exacerbates cellular inflammation and chemokine production. We demonstrate increased CCL2 and ACKR2 mRNA expression in post-mortem human brain, whereby ACKR2 mRNA levels correlated with later times post-TBI. This data is consistent with the transient upregulation of ACKR2 observed in mouse brain after closed head injury (CHI). As compared to WT animals, ACKR2-/- mice showed a higher mortality rate after CHI, while the neurological outcome in surviving mice was similar. At day 1 post-injury, ACKR2-/- mice displayed aggravated lesion volume and no differences in CCL2 expression and macrophage recruitment relative to WT mice. Reciprocal regulation of ACKR2 and CCL2 expression was explored in cultured astrocytes, which are recognized as the major source of CCL2 and also express ACKR2. ACKR2 mRNA increased as early as 2 hours after an inflammatory challenge in WT astrocytes. As expected, CCL2 expression also dramatically increased at 4 hours in WT astrocytes but was significantly lower in ACKR2-/- astrocytes, possibly indicating a co-regulation of CCL2 and ACKR2 in these cells. Conversely, in vivo, CCL2 mRNA/protein levels were increased similarly in ACKR2-/- and WT brains at 4 and 12 hours after CHI, in line with the lack of differences in cerebral macrophage recruitment and neurological recovery. In conclusion, ACKR2 is induced after TBI and has a significant impact on mortality and lesion development acutely following CHI, while its role in chemokine expression, macrophage activation, brain pathology, and neurological recovery at later time-points is minor. Concordant to evidence in multiple sclerosis experimental models, our data corroborate a distinct role for ACKR2 in cerebral inflammatory processes compared to its reported functions in peripheral tissues.
- Published
- 2017
23. Prolonged intrathecal release of soluble Fas following severe traumatic brain injury in humans
- Author
-
Otmar Trentz, Maria-Cristina Morganti-Kossmann, Andreas Marx, Philipp M. Lenzlinger, and Thomas Kossmann
- Subjects
Adult ,Male ,medicine.medical_specialty ,Programmed cell death ,Fas Ligand Protein ,Adolescent ,medicine.medical_treatment ,Immunology ,Apoptosis ,Neopterin ,Fas ligand ,Cerebrospinal fluid ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,fas Receptor ,CSF albumin ,Aged ,Membrane Glycoproteins ,Tumor Necrosis Factor-alpha ,business.industry ,Receptors, Interleukin-2 ,Middle Aged ,Fas receptor ,Endocrinology ,Cytokine ,Solubility ,Neurology ,Blood-Brain Barrier ,Brain Injuries ,Phosphopyruvate Hydratase ,Encephalitis ,Female ,Tumor necrosis factor alpha ,Neurology (clinical) ,business ,Biomarkers - Abstract
The mechanisms underlying cell death following traumatic brain injury (TBI) are not fully understood. Apoptosis is believed to be one mechanism contributing to a marked and prolonged neuronal cell loss following TBI. Recent data suggest a role for Fas (APO-1, CD95), a type I transmembrane receptor glycoprotein of the nerve growth factor/tumor necrosis factor superfamily, and its ligand (Fas ligand, FasL) in apoptotic events in the central nervous system. A truncated form of the Fas receptor, soluble Fas (sFas) may indicate activation of the Fas/FasL system and act as a negative feedback mechanism, thereby inhibiting Fas mediated apoptosis. Soluble Fas was measured in cerebrospinal fluid (CSF) and serum of 10 patients with severe TBI (GCS< or =8) for up to 15 days post-trauma. No sFas was detected in CSF samples from patients without neurological pathologies. Conversely, after TBI 118 out of 120 CSF samples showed elevated sFas concentrations ranging from 56 to 4327 mU/ml. Paired serum samples showed above normal (8.5 U/ml) sFas concentrations in 5 of 10 patients. Serum levels of sFas were always higher than CSF levels. However, there was no correlation between concentrations measured in CSF and in serum (r(2)=0.078, p=0.02), suggesting that the concentrations in the two compartments are independently regulated. Also, no correlation was found between sFas in CSF and blood brain barrier (BBB) dysfunction as assessed by the albumin CSF/serum quotient (Q(A)), and concentrations of the cytotoxic cytokine tumor necrosis factor-alpha in CSF, respectively. Furthermore, there was no correlation with two markers of immune activation (soluble interleukin-2 receptor and neopterin) in CSF. Maximal CSF levels of sFas correlated significantly (r(2)=0.8191, p
- Published
- 2002
24. Traumatic brain injury induces elevation of Co in the human brain
- Author
-
Ashley I. Bush, Dominic J. Hare, Qiao-Xin Li, Alison Conquest, Tony Frugier, Colin L. Masters, Maria-Cristina Morganti-Kossmann, Catriona McLean, Monica Lind, and Blaine R. Roberts
- Subjects
Adult ,Male ,medicine.medical_specialty ,Injury control ,Adolescent ,Traumatic brain injury ,Biophysics ,Poison control ,Autopsy ,Biochemistry ,Biomaterials ,Cohort Studies ,Young Adult ,Neuroimaging ,Internal medicine ,Metals, Heavy ,medicine ,Humans ,Aged ,Brain Chemistry ,medicine.diagnostic_test ,business.industry ,Head injury ,Metals and Alloys ,Human brain ,Cobalt ,Middle Aged ,medicine.disease ,nervous system diseases ,Endocrinology ,medicine.anatomical_structure ,nervous system ,Chemistry (miscellaneous) ,Positron emission tomography ,Brain Injuries ,Female ,business - Abstract
Traumatic brain injury (TBI) is the most common cause of death and disability in young adults, yet the molecular mechanisms that follow TBI are poorly understood. We previously reported a perturbation in iron (Fe) levels following TBI. Here we report that the distribution of cobalt (Co) is modulated in post-mortem human brain following injury. We also investigated how the distribution of other biologically relevant elements changes in TBI. Cobalt is increased due to TBI while copper (Cu), magnesium (Mg), manganese (Mn), phosphorus (P), potassium (K), rubidium (Rb), selenium (Se) and zinc (Zn) remain unchanged. The elevated Co has important implications for positron emission tomography neuroimaging. This is the first demonstration of the accumulation of Co in injured tissue explaining the previous utility of (55)Co-PET imaging in TBI.
- Published
- 2014
25. Anti-lysophosphatidic acid antibodies improve traumatic brain injury outcomes
- Author
-
Jonathan M Wojciak, Robert L. Medcalf, Peter J Crack, Leigh A. Johnston, Moses Zhang, Jonathan K. Fleming, Alison Conquest, Maria Daglas, Roger A. Sabbadini, Maithili Sashindranath, Andrew J. Morris, Yona Goldshmit, David W. Wright, Alice Pébay, Maria Cristina Morganti-Kossmann, and Ila P. Karve
- Subjects
Nervous system ,Male ,Pathology ,Neurology ,Human cerebrospinal fluid ,chemistry.chemical_compound ,Mice ,Control cortical impact ,Cerebrospinal fluid ,Traumatic brain injury ,Lysophosphatidic acid ,Single-Blind Method ,Aged, 80 and over ,General Neuroscience ,Middle Aged ,3. Good health ,medicine.anatomical_structure ,Cytokines ,lipids (amino acids, peptides, and proteins) ,Female ,medicine.symptom ,Autotaxin ,biological phenomena, cell phenomena, and immunity ,Adult ,medicine.medical_specialty ,Immunology ,Short Report ,Brain damage ,Neuroprotection ,Cellular and Molecular Neuroscience ,Young Adult ,Magnetic resonance imaging ,medicine ,Animals ,Humans ,Immunologic Factors ,Glasgow Coma Scale ,IL-6 ,business.industry ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,chemistry ,Anti-LPA antibody ,Brain Injuries ,Immunoglobulin G ,Lysophospholipids ,business - Abstract
Background Lysophosphatidic acid (LPA) is a bioactive phospholipid with a potentially causative role in neurotrauma. Blocking LPA signaling with the LPA-directed monoclonal antibody B3/Lpathomab is neuroprotective in the mouse spinal cord following injury. Findings Here we investigated the use of this agent in treatment of secondary brain damage consequent to traumatic brain injury (TBI). LPA was elevated in cerebrospinal fluid (CSF) of patients with TBI compared to controls. LPA levels were also elevated in a mouse controlled cortical impact (CCI) model of TBI and B3 significantly reduced lesion volume by both histological and MRI assessments. Diminished tissue damage coincided with lower brain IL-6 levels and improvement in functional outcomes. Conclusions This study presents a novel therapeutic approach for the treatment of TBI by blocking extracellular LPA signaling to minimize secondary brain damage and neurological dysfunction.
- Published
- 2014
26. Post-Traumatic hypoxia is associated with prolonged cerebral cytokine production, higher serum biomarker levels, and poor outcome in patients with severe traumatic brain injury
- Author
-
Phuong Nguyen, Maria Cristina Morganti-Kossmann, Nicole Bye, Jeffrey V. Rosenfeld, Edwin BingBing Yan, D. Agyapomaa, Eldho Paul, Laveniya Satgunaseelan, and Thomas Kossmann
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pathology ,Adolescent ,Traumatic brain injury ,Serum albumin ,Enzyme-Linked Immunosorbent Assay ,Blood–brain barrier ,Gastroenterology ,Young Adult ,Cerebrospinal fluid ,Internal medicine ,medicine ,Humans ,Glasgow Coma Scale ,Hypoxia, Brain ,Neuroinflammation ,biology ,business.industry ,Glasgow Outcome Scale ,Recovery of Function ,Original Articles ,Hypoxia (medical) ,Middle Aged ,medicine.disease ,Prognosis ,medicine.anatomical_structure ,nervous system ,Blood-Brain Barrier ,Brain Injuries ,biology.protein ,Cytokines ,Female ,Neurology (clinical) ,Human medicine ,medicine.symptom ,business ,Biomarkers - Abstract
Secondary hypoxia is a known contributor to adverse outcomes in patients with traumatic brain injury (TBI). Based on the evidence that hypoxia and TBI in isolation induce neuroinflammation, we investigated whether TBI combined with hypoxia enhances cerebral cytokine production. We also explored whether increased concentrations of injury biomarkers discriminate between hypoxic (Hx) and normoxic (Nx) patients, correlate to worse outcome, and depend on blood-brain barrier (BBB) dysfunction. Forty-two TBI patients with Glasgow Coma Scale
- Published
- 2014
27. Regulation of chemokines and chemokine receptors after experimental closed head injury
- Author
-
Karin Kariya, Ido Yatsiv, Viviane I. Otto, Otmar Trentz, Esther Shohami, Thomas Kossmann, Philip F. Stahel, Mario Rancan, Maria Cristina Morganti-Kossmann, and Hans-Pietro Eugster
- Subjects
medicine.medical_specialty ,Chemokine ,Receptors, CCR5 ,Chemokine CXCL2 ,Biology ,Receptors, Interleukin-8B ,Mice ,Chemokine receptor ,Head Injuries, Closed ,Internal medicine ,medicine ,Animals ,CXC chemokine receptors ,Chemokine CCL4 ,Receptor ,Lymphotoxin-alpha ,Macrophage inflammatory protein ,Chemokine CCL3 ,Cerebral Cortex ,Mice, Knockout ,Microglia ,Tumor Necrosis Factor-alpha ,Macrophages ,Monokines ,General Neuroscience ,Macrophage Inflammatory Proteins ,Up-Regulation ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,Gene Expression Regulation ,Astrocytes ,Immunology ,biology.protein ,Encephalitis ,Neuroglia ,Receptors, Chemokine ,Tumor necrosis factor alpha ,Chemokines - Abstract
The expression of the chemokines macrophage inflammatory protein (MIP)-2 and MIP-1alpha and of their receptors CXCR2 and CCR5 was assessed in wild type (WT) and TNF/lymphotoxin-alpha knockout (TNF/LT-alpha-/-) mice subjected to closed head injury (CHI). At 4 h after trauma intracerebral MIP-2 and MIP-1alpha levels were increased in both groups with MIP-2 concentrations being significantly higher in WT than in TNF/LT-alpha-/- animals (p < 0.05). Thereafter, MIP-2 production declined rapidly, whereas MIP-1alpha remained elevated for 7 days. Expression of CXCR2 was confined to astrocytes and increased dramatically within 24 h in both mouse types. Contrarily, CCR5 expression remained constitutively low and was mainly localized to microglia. These results show that after CHI, chemokines and their receptors are regulated differentially and with independent kinetics.
- Published
- 2001
28. The Duality of the Inflammatory Response to Traumatic Brain Injury
- Author
-
Helmut L. Laurer, Maria-Cristina Morganti-Kossmann, Philipp M. Lenzlinger, and Tracy K. McIntosh
- Subjects
Inflammation ,Microglia ,business.industry ,Traumatic brain injury ,Central nervous system ,Neuroscience (miscellaneous) ,Brain damage ,Blood–brain barrier ,medicine.disease ,Models, Biological ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Traumatic injury ,Neurology ,Brain Injuries ,Immunology ,medicine ,Animals ,Cytokines ,Humans ,Tumor necrosis factor alpha ,medicine.symptom ,business - Abstract
One and a half to two million people sustain a traumatic brain injury (TBI) in the US each year, of which approx 70,000-90,000 will suffer from long-term disability with dramatic impacts on their own and their families' lives and enormous socio-economic costs. Brain damage following traumatic injury is a result of direct (immediate mechanical disruption of brain tissue, or primary injury) and indirect (secondary or delayed) mechanisms. These secondary mechanisms involve the initiation of an acute inflammatory response, including breakdown of the blood-brain barrier (BBB), edema formation and swelling, infiltration of peripheral blood cells and activation of resident immunocompetent cells, as well as the intrathecal release of numerous immune mediators such as interleukins and chemotactic factors. An overview over the inflammatory response to trauma as observed in clinical and in experimental TBI is presented in this review. The possibly harmful/beneficial sequelae of post-traumatic inflammation in the central nervous system (CNS) are discussed using three model mediators of inflammation in the brain, tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and transforming growth factor-beta (TGF-beta). While the former two may act as important mediators for the initiation and the support of post-traumatic inflammation, thus causing additional cell death and neurologic dysfunction, they may also pave the way for reparative processes. TGF-beta, on the other hand, is a potent anti-inflammatory agent, which may also have some deleterious long-term effects in the injured brain. The implications of this duality of the post-traumatic inflammatory response for the treatment of brain-injured patients using anti-inflammatory strategies are discussed.
- Published
- 2001
29. Intracerebral complement C5a receptor (CD88) expression is regulated by TNF and lymphotoxin-α following closed head injury in mice
- Author
-
Otmar Trentz, Philip F. Stahel, Karin Kariya, Esther Shohami, Maria Cristina Morganti-Kossmann, Hans-Pietro Eugster, Scott R. Barnum, and Thomas Kossmann
- Subjects
Lymphotoxin alpha ,medicine.medical_specialty ,Immunology ,Gene Expression ,Inflammation ,Biology ,C5a receptor ,Mice ,Antigens, CD ,Head Injuries, Closed ,Internal medicine ,Gene expression ,medicine ,Animals ,Immunology and Allergy ,RNA, Messenger ,Receptor ,Lymphotoxin-alpha ,Receptor, Anaphylatoxin C5a ,In Situ Hybridization ,Brain Chemistry ,Mice, Knockout ,Tumor Necrosis Factor-alpha ,Receptors, Complement ,Mice, Inbred C57BL ,Endocrinology ,Lymphotoxin ,Neurology ,Knockout mouse ,Tumor necrosis factor alpha ,Neurology (clinical) ,medicine.symptom - Abstract
The anaphylatoxin C5a is a potent mediator of inflammation in the CNS. We analyzed the intracerebral expression of the C5a receptor (C5aR) in a model of closed head injury (CHI) in mice. Up-regulation of C5aR mRNA and protein expression was observed mainly on neurons in sham-operated and head-injured wild-type mice at 24 h. In contrast, in TNF/lymphotoxin-alpha knockout mice, the intracerebral C5aR expression remained at low constitutive levels after sham operation, whereas it strongly increased in response to trauma between 24 and 72 h. Interestingly, by 7 days after CHI, the intrathecal C5aR expression was clearly attenuated in the knockout animals. These data show that the posttraumatic neuronal expression of the C5aR is, at least in part, regulated by TNF and lymphotoxin-alpha at 7 days after trauma.
- Published
- 2000
30. The role of inflammation in neurologic disease
- Author
-
Philip F. Stahel, Viviane I. Otto, Thomas Kossmann, and Maria Cristina Morganti-Kossmann
- Subjects
medicine.anatomical_structure ,business.industry ,Central nervous system ,Immunology ,medicine ,Inflammation ,medicine.symptom ,Neurologic disease ,Critical Care and Intensive Care Medicine ,CNS tissue ,business ,Homeostasis - Abstract
The central nervous system (CNS) requires an intact and peculiar environment in order to function properly. This homeostasis is maintained by the blood-brain barrier, which separates the CNS from the peripheral circulation. The ability of the CNS tissue to counteract the pathogenic effect of infecti
- Published
- 2000
31. sICAM-1 and TNF-? induce MIP-2 with distinct kinetics in astrocytes and brain microvascular endothelial cells
- Author
-
Sergio M. Gloor, Otmar Trentz, Maria Cristina Morganti-Kossmann, Viviane I. Otto, Thomas Kossmann, and Ulrike E. Heinzel-Pleines
- Subjects
Chemokine ,Pathology ,medicine.medical_specialty ,biology ,Cell adhesion molecule ,Interleukin ,Intercellular adhesion molecule ,Cellular and Molecular Neuroscience ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,biology.protein ,medicine ,Tumor necrosis factor alpha ,Receptor ,Macrophage inflammatory protein - Abstract
The dysfunction of the blood-brain barrier (BBB) occurring after traumatic brain injury (TBI) is mediated by intracerebral neutrophil accumulation, chemokine release (e.g., interleukin (IL)-8) and upregulation of adhesion molecules (e.g., intercellular adhesion molecule (ICAM)-1). In patients with severe TBI, we previously found that elevated cerebrospinal fluid (CSF) IL-8 and soluble (s)ICAM-1 correlate with BBB dysfunction, and this prompted us to concomitantly monitor IL-8, sICAM-1 and their stimulator tumor necrosis factor (TNF)-alpha in CSF. Potential mechanisms for upregulation of the IL-8 analogue, murine macrophage inflammatory protein (MIP)-2, and sICAM-1 at the BBB were studied using cultured mouse astrocytes and brain microvascular endothelial cells (MVEC). In CSF of seven patients, IL-8 and sICAM-1 were elevated for 19 days after severe TBI, whereas TNF-alpha exceeded normal values on 9 days. Stimulation of MVEC and astrocytes with TNF-alpha simultaneously induced the release of MIP-2 reaching saturation by 4-8 hr and of sICAM-1 increasing continuously from 2-4 hr to 12 hr. Augmented sICAM-1 production correlated with enhanced membrane-bound (m)ICAM-1 expression in both cell types (r(s) = 0.96 and 0.90, P < 0.0001), but was markedly higher in astrocytes. The release of sICAM-1 was not influenced by IL-8 or MIP-2, although astrocytes and MVEC expressed the IL-8/MIP-2 receptor (CXCR-2) as determined by FACS analysis. Instead, we found that sICAM-1 strongly induced MIP-2 secretion by both cell types with kinetics differing from those evoked by TNF-alpha. If added together, sICAM-1 and TNF-alpha synergistically induced MIP-2 production suggesting the involvement of two different pathways for MIP-2 regulation.
- Published
- 2000
32. IL-10 levels in cerebrospinal fluid and serum of patients with severe traumatic brain injury: relationship to IL-6, TNF-α, TGF-β1 and blood–brain barrier function
- Author
-
Maria Cristina Morganti-Kossmann, Helen Joller, Esther Csuka, Thomas Kossmann, Philipp M. Lenzlinger, and Otmar Trentz
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Traumatic brain injury ,medicine.medical_treatment ,Immunology ,Inflammation ,Blood–brain barrier ,Cerebrospinal fluid ,Transforming Growth Factor beta ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Interleukin 6 ,Aged ,biology ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,business.industry ,Middle Aged ,medicine.disease ,Interleukin-10 ,Interleukin 10 ,Cytokine ,Endocrinology ,medicine.anatomical_structure ,Neurology ,Blood-Brain Barrier ,Brain Injuries ,biology.protein ,Female ,Tumor necrosis factor alpha ,Neurology (clinical) ,medicine.symptom ,business - Abstract
Controlling the extent of inflammatory responses following brain injury may be beneficial since posttraumatic intracranial inflammation has been associated with adverse outcome. In order to elucidate the potential role of anti-inflammatory mediators, the production of interleukin-10 (IL-10) was monitored in paired cerebrospinal fluid (CSF) and serum of 28 patients with severe traumatic brain injury (TBI) and compared to control samples. The pattern of IL-10 was analyzed with respect to the patterns of IL-6, tumor necrosis factor-alpha (TNF-alpha) and transforming growth factor-beta1 (TGF-beta1) in both fluids during a time period of up to 22 days. In parallel, the function/dysfunction of the blood-brain barrier (BBB) was monitored using the CSF-/serum-albumin quotient (Q(A)) and compared to intrathecal cytokine levels. Mean IL-10 concentration in CSF was elevated in 26 out of 28 TBI patients (range: 1.3-41.7 pg/ml) compared to controls (cut-off: 1.06 pg/ml), whereas only seven patients had elevated mean IL-10 concentration in serum (range: 5.4-23 pg/ml; cut-off: 5.14 pg/ml). The time course of IL-10 was similar in both fluids, showing a peak during the first days and a second, lower rise in the second week. Intrathecal IL-10 synthesis is hypothesized since CSF-IL-10 levels exceeded serum-IL-10 levels in most of the patients, IL-10-index (CSF/serum-IL-10/QA) was elevated in 23 individuals, and elevation of CSF-IL-10 showed to be independent from severe BBB dysfunction. Neither CSF nor serum IL-10 values correlated with the dysfunction of the BBB. IL-10, IL-6 and TGF-beta1 showed similar patterns in CSF over time, whereas rises of TNF-alpha corresponded to declines of IL-10 levels. Our results suggest that IL-10 is predominantly induced intrathecally after severe TBI where it may downregulate inflammatory events following traumatic brain damage.
- Published
- 1999
33. Experimental Axonal Injury Triggers Interleukin-6 mRNA, Protein Synthesis and Release into Cerebrospinal Fluid
- Author
-
Maria Cristina Morganti-Kossmann, Otmar Trentz, Rainer Probstmeier, Thomas Kossmann, Volkmar Hans, Hans-Georg Imhof, and Philipp M. Lenzlinger
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Nerve Crush ,medicine.medical_treatment ,In situ hybridization ,Hippocampal formation ,030218 nuclear medicine & medical imaging ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,medicine ,Animals ,RNA, Messenger ,Axon ,In Situ Hybridization ,biology ,Interleukin-6 ,Diffuse axonal injury ,medicine.disease ,Immunohistochemistry ,Axons ,Rats ,medicine.anatomical_structure ,Cytokine ,Neurology ,biology.protein ,Biological Assay ,Neurology (clinical) ,Neuron ,NeuN ,Cardiology and Cardiovascular Medicine ,030217 neurology & neurosurgery - Abstract
Diffuse axonal injury is a frequent pathologic sequel of head trauma, which, despite its devastating consequences for the patients, remains to be fully elucidated. Here we studied the release of interleukin-6 (IL-6) into CSF and serum, as well as the expression of IL-6 messenger ribonucleic acid (mRNA) and protein in a weight drop model of axonal injury in the rat. The IL-6 activity was elevated in CSF within 1 hour and peaked between 2 and 4 hours, reaching maximal values of 82,108 pg/mL, and returned to control values after 24 hours. In serum, the levels of IL-6 remained below increased CSF levels and did not exceed 393 pg/mL. In situ hybridization demonstrated augmented IL-6 mRNA expression in several regions including cortical pyramidal cells, neurons in thalamic nuclei, and macrophages in the basal subarachnoid spaces. A weak constitutive expression of IL-6 protein was shown by immunohistochemical study in control brain. After injury, IL-6 increased at 1 hour and remained elevated through the first 24 hours, returning to normal afterward. Most cells producing IL-6 were cortical, thalamic, and hippocampal neurons as confirmed by staining for the neuronal marker NeuN. These results extend our previous studies showing IL-6 production in the cerebrospinal fluid of patients with severe head trauma and demonstrate that neurons are the main source of IL-6 after experimental axonal injury.
- Published
- 1999
34. Glutamate and Taurine Are Increased in Ventricular Cerebrospinal Fluid of Severely Brain-Injured Patients
- Author
-
Philipp M. Lenzlinger, Maria Cristina Morganti-Kossmann, Thomas Kossmann, John F. Stover, Reto Stocker, and Oliver S. Kempski
- Subjects
Adult ,Male ,Taurine ,Pathology ,medicine.medical_specialty ,Glutamic Acid ,Brain Edema ,Brain damage ,Severity of Illness Index ,Cerebral Ventricles ,Central nervous system disease ,chemistry.chemical_compound ,Cerebrospinal fluid ,In vivo ,medicine ,Extracellular ,Humans ,Glasgow Coma Scale ,Longitudinal Studies ,Brain Concussion ,Aged ,Monitoring, Physiologic ,Analysis of Variance ,business.industry ,Glutamate receptor ,Middle Aged ,medicine.disease ,Pathophysiology ,chemistry ,Brain Injuries ,Disease Progression ,Female ,Neurology (clinical) ,medicine.symptom ,business ,Biomarkers - Abstract
Glutamate contributes to secondary brain damage, resulting in cell swelling and brain edema. Under in vitro conditions, increased extracellular levels of the amino acid taurine reflect glutamate-induced osmotic cell swelling. In vivo, increases in cerebrospinal fluid (CSF) taurine could, therefore, unmask glutamate-mediated cytotoxic edema formation and possibly differentiate it from vasogenic edema. To test this hypothesis, ventricular CSF glutamate and taurine levels were measured in 28 severely brain-injured patients on days 1, 5, and 14 after trauma. Posttraumatic changes in CSF amino acids were investigated in regard to extent of tissue damage and alterations in brain edema as estimated by computerized tomography. On day 1, CSF glutamate and taurine levels were significantly increased in patients with subdural or epidural hematomas (8+/-0.8/71+/-12 microM), contusions (21+/-4.1/122+/-18 microM), and generalized brain edema (13+/-3.2/80+/-15 microM) compared to lumbar control CSF (1.3+/-0.1/12+/-1 microM; p0.001). CSF amino acids, however, did not reflect edema formation and resolution as estimated by computerized tomography. CSF taurine correlated positively with glutamate, eventually depicting glutamate-induced cell swelling. However, parallel neuronal release of taurine with its inhibitory function cannot be excluded. Thus, the sensitivity of taurine in unmasking cytotoxic edema formation is weakened by the inability in defining its origin and function under the conditions chosen in the present study. Overall, persisting pathologic ventricular CSF glutamate and taurine levels are highly suggestive of ongoing glial and neuronal impairment in humans following severe traumatic brain injury.
- Published
- 1999
35. Thiopental and midazolam do not seem to impede metabolism of glutamate in brain-injured patients
- Author
-
John F. Stover, Reto Stocker, Oliver S. Kempski, Maria Cristina Morganti-Kossmann, Ulrike E. Pleines, and Thomas Kossmann
- Subjects
Adult ,Adolescent ,medicine.drug_class ,Glutamine ,Midazolam ,Glutamic Acid ,Pharmacology ,Cerebral Ventricles ,Detoxification ,Serine ,medicine ,Humans ,Hypnotics and Sedatives ,Thiopental ,Aged ,Alanine ,Thiopental Sodium ,business.industry ,Glutamate receptor ,Metabolism ,Middle Aged ,Biochemistry ,Barbiturate ,Brain Injuries ,Toxicity ,Lactates ,business - Abstract
Increased extracellular glutamate levels are related to glial and neuronal damage. Glutamate-mediated toxicity is limited by glial uptake and metabolic transformation of glutamate to glutamine and the energetic compounds alanine and lactate which are utilized by surrounding neurons. Under in vitro conditions, barbiturates have been shown to reduce glutamate uptake and its further metabolism, possibly impeding metabolic coupling between astrocytes and neurons. The aims were to investigate if under clinical conditions, the barbiturate thiopental reduces important detoxification of glutamate, resulting in lower CSF glutamine, alanine and lactate levels as opposed to patients receiving midazolam. During long-term administration of thiopental and midazolam, pathologically elevated ventricular CSF glutamate levels were associated with significantly increased glutamine and alanine levels up to 14 days after trauma. CSF lactate, however, remained normal. These data suggest that long-term administration of thiopental and midazolam under clinical conditions does not impede enzymatic activities responsible for detoxification and metabolism of glutamate.
- Published
- 1999
36. Thiopental in CSF and Serum Correlates with Prolonged Loss of Cortical Activity
- Author
-
Philipp M. Lenzlinger, Reto Stocker, Otmar Trentz, Hans-Georg Imhof, Maria Cristina Morganti-Kossmann, John F. Stover, and Thomas Kossmann
- Subjects
Adult ,Male ,Time Factors ,Adolescent ,Critical Care ,Intracranial Pressure ,Traumatic brain injury ,medicine.drug_class ,Neurological disorder ,Central nervous system disease ,Cerebrospinal fluid ,Pharmacokinetics ,medicine ,Humans ,Hypnotics and Sedatives ,Thiopental ,Infusions, Intravenous ,Coma ,Thiopental Sodium ,business.industry ,Electroencephalography ,Middle Aged ,medicine.disease ,Neurology ,Barbiturate ,Brain Injuries ,Anesthesia ,Female ,Neurology (clinical) ,medicine.symptom ,business - Abstract
Barbiturate coma is initiated in brain-injured patients whenever elevated intracranial pressure remains unresponsive to other therapeutical strategies. However, barbiturates alter cortical activity resulting in difficulties in clinical evaluation. Therefore, we investigated the impact of long-term thiopental administration on responsiveness to exteroceptive stimuli in relation to pharmacokinetics of thiopental in CSF and serum. Long-term infusion increases thiopental levels which remain elevated for 6 and 9 days in CSF and serum, respectively, after termination of its administration. Prolonged unresponsiveness to exteroceptive stimuli correlates with persisting thiopental in CSF and serum. Thus, quantitative analysis of thiopental in serum becomes indispensable in predicting the length of drug-induced neurological impairment and in avoiding misinterpretation of the neurological status.
- Published
- 1998
37. Neurotransmitters in cerebrospinal fluid reflect pathological activity
- Author
-
Oliver Kempski, Ulrike E. Pleines, Maria Cristina Morganti-Kossmann, Thomas Kossmann, John F. Stover, and K. Lowitzsch
- Subjects
Adult ,Male ,medicine.medical_specialty ,Taurine ,Pathology ,Glutamine ,Clinical Biochemistry ,Glycine ,Glutamic Acid ,Blood–brain barrier ,Biochemistry ,Myelopathy ,chemistry.chemical_compound ,Cerebrospinal fluid ,Central Nervous System Diseases ,Internal medicine ,Serine ,medicine ,Viral meningitis ,Humans ,Aged ,Aspartic Acid ,Neurotransmitter Agents ,L-Lactate Dehydrogenase ,business.industry ,Multiple sclerosis ,Glutamate receptor ,General Medicine ,Middle Aged ,medicine.disease ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Blood-Brain Barrier ,Female ,Asparagine ,business - Abstract
The excitatory transmitters glutamate and aspartate become toxic whenever their extracellular levels are increased because of neuronal, glial and endothelial impairment. Taurine, a volume-regulating amino acid, is released upon excitotoxin-induced cell swelling. Our aim was to investigate if glutamate and aspartate in cerebrospinal fluid (CSF) reveal neuropathology in neurological patients, and if taurine unmasks glutamate-mediated toxicity. Glutamate and aspartate are doubled in viral meningitis, acute multiple sclerosis (MS) and myelopathy compared with control subjects and patients with peripheral facial nerve palsy. These levels do not coincide with a disturbed blood-brain barrier, as estimated by the albumin ratio, are independent of their precursors (glutamine, asparagine) and are not associated with cell lysis. Taurine is significantly increased in meningitis, acute MS, and myelopathy, suggesting glutamate-mediated toxicity. Analysis of transmitters in lumbar CSF can be used to identify patients with cerebral and spinal pathology who might benefit from specific receptor-modulating agents.
- Published
- 1997
38. Neurochemical alterations and current pharmacological strategies in the treatment of traumatic brain injury
- Author
-
Maria Cristina Morganti-Kossmann, J. F. Stover, Reto Stocker, Philipp M. Lenzlinger, Thomas Kossmann, and Otmar Trentz
- Subjects
medicine.medical_specialty ,Injury control ,business.industry ,Traumatic brain injury ,Poison control ,Brain damage ,medicine.disease ,Craniocerebral trauma ,Neurochemical ,Intervention (counseling) ,Emergency Medicine ,Medicine ,Orthopedics and Sports Medicine ,Surgery ,medicine.symptom ,business ,Psychiatry ,Neuroscience ,Clinical evaluation - Abstract
In recent years, our knowledge concerning pathophysiological changes in brain metabolism after traumatic brain injury (TBI) has greatly expanded. This, in turn, has enabled the development of specific pharmacological strategies for the supplementary treatment of brain-injured patients with the aim of reducing secondary brain damage. The present article focuses on the pathophysiology of TBI and the possibilities for pharmacological intervention. While some of the substances reviewed are presently used in the treatment of TBI, others are under experimental and clinical evaluation at different stages.
- Published
- 1997
39. Interleukin-8 Released into the Cerebrospinal Fluid after Brain Injury is Associated with Blood–Brain Barrier Dysfunction and Nerve Growth Factor Production
- Author
-
Rolf W. Dubs, Heinz Redl, Guenter Schlag, Philipp M. Lenzlinger, Thomas Kossmann, Otmar Trentz, Philip F. Stahel, and Maria Cristina Morganti-Kossmann
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Nerve growth factor production ,Blood–brain barrier ,030218 nuclear medicine & medical imaging ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Internal medicine ,Animals ,Humans ,Medicine ,Nerve Growth Factors ,Interleukin 8 ,Interleukin 6 ,Cells, Cultured ,Aged ,biology ,business.industry ,Interleukin-8 ,Interleukin ,Cerebrospinal Fluid Proteins ,Middle Aged ,Recombinant Proteins ,Cytokine ,medicine.anatomical_structure ,Nerve growth factor ,Endocrinology ,Gene Expression Regulation ,Neurology ,Blood-Brain Barrier ,Astrocytes ,Brain Injuries ,Immunology ,biology.protein ,Female ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery - Abstract
Interleukin (IL) 8 was measured in CSF of 14 patients with severe traumatic brain injury. IL-8 levels were significantly higher in CSF (up to 8,000 pg/ml) than serum (up to 2,400 pg/ml) (p < 0.05), suggesting intrathecal production. Maximal IL-8 values in CSF correlated with a severe dysfunction of the blood–brain barrier. Nerve growth factor (NGF) was detected in CSF of 7 of 14 patients (range of maximal NGF: 62–12,130 pg/ml). IL-8 concentrations were significantly higher in these patients than in those without NGF (p < 0.01). CSF containing high IL-8 (3,800–7,900 pg/ml) induced greater NGF production in cultured astrocytes (202–434 pg/ml) than samples with low IL-8 (600–1,000 pg/ml), which showed a smaller NGF increase (0–165 pg/ml). Anti-IL-8 antibodies strongly reduced (52–100%) the release of NGF in the group of high IL-8, whereas in the group with low IL-8, this effect was lower (0–52%). The inability of anti-IL-8 antibodies to inhibit the synthesis of NGF completely may depend on cytokines like tumor necrosis factor α and IL-6 found in these CSF samples, which may act in association with IL-8. Thus, IL-8 may represent a pivotal cytokine in the pathology of brain injury.
- Published
- 1997
40. Elevated levels of the complement components C3 and factor B in ventricular cerebrospinal fluid of patients with traumatic brain injury
- Author
-
Maria Cristina Morganti-Kossmann, Philip F. Stahel, Jennifer Jones, Thomas Kossmann, and Scott R. Barnum
- Subjects
Adult ,Male ,Adolescent ,Traumatic brain injury ,Immunology ,Central nervous system ,Blood–brain barrier ,Complement factor B ,Head trauma ,Cerebrospinal fluid ,Reference Values ,medicine ,Humans ,Immunology and Allergy ,Aged ,business.industry ,Complement C3 ,Middle Aged ,medicine.disease ,Complement system ,medicine.anatomical_structure ,Neurology ,Blood-Brain Barrier ,Brain Injuries ,Anesthesia ,Alternative complement pathway ,Female ,Neurology (clinical) ,business ,Complement Factor B - Abstract
Immunological events occurring in the central nervous system (CNS) as a result of head trauma are largely unexplored. We report here that the levels of the alternative pathway complement proteins C3 and factor B are elevated in the cerebrospinal fluid (CSF) of head-injured patients. C3 and factor B indices suggest that changes in C3 and factor B levels in CSF are most likely due to altered blood-brain barrier integrity and not to intrathecal synthesis. These data demonstrate, for the first time, elevated levels of complement proteins in CSF of patients with severe traumatic brain injury. Elevated complement levels in brain injury may contribute to secondary damage.
- Published
- 1997
41. Characterising effects of impact velocity on brain and behaviour in a model of diffuse traumatic axonal injury
- Author
-
Duwage Dasuni Sathsara Alwis, Ramesh Rajan, Maria Cristina Morganti-Kossmann, Edwin BingBing Yan, and Victoria P.A. Johnstone
- Subjects
Male ,Neurofilament ,Traumatic brain injury ,Acceleration ,Diffuse Axonal Injury ,Corpus Callosum ,Rats, Sprague-Dawley ,Impact velocity ,Lateral Ventricles ,medicine ,Animals ,Sensory cortex ,Neurons ,Impact acceleration ,General Neuroscience ,Diffuse axonal injury ,Biomechanics ,Somatosensory Cortex ,medicine.disease ,Biomechanical Phenomena ,Electrophysiology ,Disease Models, Animal ,medicine.anatomical_structure ,Rotarod Performance Test ,Psychology ,Neuroscience - Abstract
The velocity of impact between an object and the human head is a critical factor influencing brain injury outcomes but has not been explored in any detail in animal models. Here we provide a comprehensive overview of the interplay between impact velocity and injury severity in a well-established weight-drop impact acceleration (WDIA) model of diffuse brain injury in rodents. We modified the standard WDIA model to produce impact velocities of 5.4, 5.85 and 6.15 m/s while keeping constant the weight and the drop height. Gradations in impact velocity produced progressive degrees of injury severity measured behaviourally, electrophysiologically and anatomically, with the former two methods showing greater sensitivity to changes in impact velocity. There were impact velocity-dependent reductions in sensorimotor performance and in cortical depth-related depression of sensory cortex responses; however axonal injury (demonstrated by immunohistochemistry for β-amyloid precursor protein and neurofilament heavy-chain) was discernible only at the highest impact velocity. We conclude that the WDIA model is capable of producing graded axonal injury in a repeatable manner, and as such will prove useful in the study of the biomechanics, pathophysiology and potential treatment of diffuse axonal injury.
- Published
- 2013
42. The Role of Markers of Inflammation in Traumatic Brain Injury
- Author
-
Maria Cristina Morganti-Kossmann and Thomas M. Woodcock
- Subjects
Chemokine ,Traumatic brain injury ,medicine.medical_treatment ,Inflammation ,chemokines ,Brain damage ,Review Article ,lcsh:RC346-429 ,Immune system ,Medicine ,Neuroinflammation ,lcsh:Neurology. Diseases of the nervous system ,biology ,Microglia ,business.industry ,traumatic brain injury ,biomarkers ,medicine.disease ,cytokines ,medicine.anatomical_structure ,Cytokine ,Neurology ,inflammation ,Immunology ,biology.protein ,Neurology (clinical) ,medicine.symptom ,business ,Neuroscience - Abstract
Within minutes of a traumatic impact, a robust inflammatory response is elicited in the injured brain. The complexity of this post-traumatic squeal involves a cellular component, comprising the activation of resident glial cells, microglia, and astrocytes, and the infiltration of blood leukocytes. The second component regards the secretion immune mediators, which can be divided into the following sub-groups: the archetypal pro-inflammatory cytokines (Interleukin-1, Tumor Necrosis Factor, Interleukin-6), the anti-inflammatory cytokines (IL-4, Interleukin-10, and TGF-beta), and the chemotactic cytokines or chemokines, which specifically drive the accumulation of parenchymal and peripheral immune cells in the injured brain region. Such mechanisms have been demonstrated in animal models, mostly in rodents, as well as in human brain. Whilst the humoral immune response is particularly pronounced in the acute phase following Traumatic brain injury (TBI), the activation of glial cells seems to be a rather prolonged effect lasting for several months. The complex interaction of cytokines and cell types installs a network of events, which subsequently intersect with adjacent pathological cascades including oxidative stress, excitotoxicity, or reparative events including angiogenesis, scarring, and neurogenesis. It is well accepted that neuroinflammation is responsible of beneficial and detrimental effects, contributing to secondary brain damage but also facilitating neurorepair. Although such mediators are clear markers of immune activation, to what extent cytokines can be defined as diagnostic factors reflecting brain injury or as predictors of long term outcome needs to be further substantiated. In clinical studies some groups reported a proportional cytokine production in either the cerebrospinal fluid or intraparenchymal tissue with initial brain damage, mortality, or poor outcome scores. However, the validity of cytokines as biomarkers is not broadly accepted. This review article will discuss the evidence from both clinical and laboratory studies exploring the validity of immune markers as a correlate to classification and outcome following TBI.
- Published
- 2013
- Full Text
- View/download PDF
43. Penetration of cefuroxime into the cerebrospinal fluid of patients with traumatic brain injury
- Author
-
Otmar Trentz, Volkmar Hans, Reto Stocker, Hans-Georg Imhof, Beda Joos, Thomas Kossmann, Maria Cristina Morganti-Kossmann, University of Zurich, and Morganti-Kossmann, M C
- Subjects
Adult ,Male ,Microbiology (medical) ,Ventriculostomy ,Adolescent ,Traumatic brain injury ,medicine.medical_treatment ,610 Medicine & health ,Blood–brain barrier ,142-005 142-005 ,2726 Microbiology (medical) ,Central nervous system disease ,Cerebrospinal fluid ,medicine ,2736 Pharmacology (medical) ,Humans ,Pharmacology (medical) ,Antibacterial agent ,Pharmacology ,Cefuroxime ,Chemotherapy ,business.industry ,2725 Infectious Diseases ,Antibiotic Prophylaxis ,medicine.disease ,Cephalosporins ,3004 Pharmacology ,Infectious Diseases ,medicine.anatomical_structure ,Blood-Brain Barrier ,Brain Injuries ,Anesthesia ,570 Life sciences ,biology ,Female ,business ,medicine.drug - Abstract
Cefuroxime levels were measured in cerebrospinal fluid (CSF) and serum of four patients with traumatic brain injury following the implantation of intraventricular catheters. The levels ranged from 0.15 to 2.03 micrograms/mL in CSF and from 1.8 to 66.9 micrograms/mL in serum. No ventriculostomy related infections were detected.
- Published
- 1996
44. Sensory cortex underpinnings of traumatic brain injury deficits
- Author
-
Maria Cristina Morganti-Kossmann, Duwage Dasuni Sathsara Alwis, Ramesh Rajan, and Edwina B Yan
- Subjects
Central Nervous System ,Male ,Anatomy and Physiology ,medicine.medical_treatment ,Poison control ,Somatosensory system ,Rats, Sprague-Dawley ,0302 clinical medicine ,Cortex (anatomy) ,Molecular Cell Biology ,Neurobiology of Disease and Regeneration ,Medicine ,Animal Management ,Neurons ,0303 health sciences ,Multidisciplinary ,Neuronal Plasticity ,Animal Behavior ,Agriculture ,Sensory Systems ,Electrophysiology ,medicine.anatomical_structure ,Cellular Types ,Research Article ,medicine.medical_specialty ,Sensory processing ,Cognitive Neuroscience ,Science ,Neurophysiology ,Sensory system ,03 medical and health sciences ,Neuroplasticity ,Animals ,Sensory cortex ,Biology ,030304 developmental biology ,business.industry ,Somatosensory Cortex ,Barrel cortex ,Animal Cognition ,Surgery ,Rats ,Brain Injuries ,Vibrissae ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Traumatic brain injury (TBI) can result in persistent sensorimotor and cognitive deficits including long-term altered sensory processing. The few animal models of sensory cortical processing effects of TBI have been limited to examination of effects immediately after TBI and only in some layers of cortex. We have now used the rat whisker tactile system and the cortex processing whisker-derived input to provide a highly detailed description of TBI-induced long-term changes in neuronal responses across the entire columnar network in primary sensory cortex. Brain injury (n = 19) was induced using an impact acceleration method and sham controls received surgery only (n = 15). Animals were tested in a range of sensorimotor behaviour tasks prior to and up to 6 weeks post-injury when there were still significant sensorimotor behaviour deficits. At 8–10 weeks post-trauma, in terminal experiments, extracellular recordings were obtained from barrel cortex neurons in response to whisker motion, including motion that mimicked whisker motion observed in awake animals undertaking different tasks. In cortex, there were lamina-specific neuronal response alterations that appeared to reflect local circuit changes. Hyper-excitation was found only in supragranular layers involved in intra-areal processing and long-range integration, and only for stimulation with complex, naturalistic whisker motion patterns and not for stimulation with simple trapezoidal whisker motion. Thus TBI induces long-term directional changes in integrative sensory cortical layers that depend on the complexity of the incoming sensory information. The nature of these changes allow predictions as to what types of sensory processes may be affected in TBI and contribute to post-trauma sensorimotor deficits.
- Published
- 2012
45. In situ detection of inflammatory mediators in post mortem human brain tissue after traumatic injury
- Author
-
Tony Frugier, Catriona McLean, David O'reilly, and Maria Cristina Morganti-Kossmann
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Adolescent ,Traumatic brain injury ,Inflammation ,Proinflammatory cytokine ,Interferon-gamma ,Young Adult ,Interferon ,Diagnosis ,medicine ,Humans ,RNA, Messenger ,Aged ,business.industry ,Tumor Necrosis Factor-alpha ,Interleukins ,Interleukin ,Brain ,Granulocyte-Macrophage Colony-Stimulating Factor ,Human brain ,Middle Aged ,medicine.disease ,Traumatic injury ,medicine.anatomical_structure ,Brain Injuries ,Immunology ,Cytokines ,Encephalitis ,Tumor necrosis factor alpha ,Female ,Neurology (clinical) ,medicine.symptom ,Inflammation Mediators ,business ,Biomarkers ,medicine.drug - Abstract
Little is known about the molecular events following severe traumatic brain injury (TBI) in humans and to date there are no efficient therapies for the treatment of patients. In this study, the first of its kind in human tissue, a total of 21 post mortem trauma brain samples were analyzed. The inflammatory response within the brain tissue was explored by measuring the expression of various inflammatory cytokines at the mRNA and protein levels. These mediators were interleukin (IL)-1beta, IL-2, IL-4, IL-6, IL-8, IL-10, tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma, and granulocyte-macrophage colony-stimulating factor (GM-CSF). This study shows for the first time in human brain tissue that 1) pro-inflammatory mediator protein levels are significantly increased in situ following acute brain injury while anti-inflammatory cytokines protein levels remain unchanged; 2) the cerebral inflammatory response begins within minutes of acute TBI, much earlier than previously thought; 3) IL-6, IL-8, TNF-alpha, and IL-1beta mRNA levels are significantly increased following injury; 4) the rise in cytokine protein level coincides with increased levels of their mRNAs suggesting that traumatic injury elicits an immediate cerebral inflammatory response. Altogether these data confirm and extend previous observations on the release of cytokines in the cerebrospinal fluid of severe TBI patients. Finally, this study highlights the need to characterize the cell source of cytokines and elucidate their mode of action.
- Published
- 2009
46. Role of chemokines in CNS health and pathology: a focus on the CCL2/CCR2 and CXCL8/CXCR2 networks
- Author
-
Maria Cristina Morganti-Kossmann, Bridgette D. Semple, and Thomas Kossmann
- Subjects
Central Nervous System ,Pathology ,medicine.medical_specialty ,CCR2 ,Chemokine ,Receptors, CCR2 ,Review Article ,Biology ,Neuroprotection ,Synaptic Transmission ,Receptors, Interleukin-8B ,Chemokine receptor ,Mice ,Central Nervous System Diseases ,medicine ,Animals ,Humans ,CXC chemokine receptors ,Chemokine CCL2 ,Macrophages ,Interleukin-8 ,CCL18 ,Chemokine activity ,CXCL2 ,Neuroprotective Agents ,Neurology ,Blood-Brain Barrier ,Immunology ,biology.protein ,Neurology (clinical) ,Chemokines ,Cardiology and Cardiovascular Medicine ,Neuroscience ,Signal Transduction - Abstract
Chemokines and their receptors have crucial roles in the trafficking of leukocytes, and are of particular interest in the context of the unique immune responses elicited in the central nervous system (CNS). The chemokine system CC ligand 2 (CCL2) with its receptor CC receptor 2 (CCR2), as well as the receptor CXCR2 and its multiple ligands CXCL1, CXCL2 and CXCL8, have been implicated in a wide range of neuropathologies, including trauma, ischemic injury and multiple sclerosis. This review aims to overview the current understanding of chemokines as mediators of leukocyte migration into the CNS under neuroinflammatory conditions. We will specifically focus on the involvement of two chemokine networks, namely CCL2/CCR2 and CXCL8/CXCR2, in promoting macrophage and neutrophil infiltration, respectively, into the lesioned parenchyma after focal traumatic brain injury. The constitutive brain expression of these chemokines and their receptors, including their recently identified roles in the modulation of neuroprotection, neurogenesis, and neurotransmission, will be discussed. In conclusion, the value of evidence obtained from the use of Ccl2- and Cxcr2-deficient mice will be reported, in the context of potential therapeutics inhibiting chemokine activity which are currently in clinical trial for various inflammatory diseases.
- Published
- 2009
47. Macrophage- and astrocyte-derived transforming growth factor beta as a mediator of central nervous system dysfunction in acquired immune deficiency syndrome
- Author
-
Uwe E. H. Mai, Maria Cristina Morganti-kossmann, J B Allen, S. E. Mergenhagen, L Ellingsworth, Sharon M. Wahl, Jan M. Orenstein, Thomas Kossmann, and Nancy McCartney-Francis
- Subjects
Male ,AIDS Dementia Complex ,medicine.medical_treatment ,Immunology ,Central nervous system ,In situ hybridization ,In Vitro Techniques ,Monocytes ,Transforming Growth Factor beta ,medicine ,Humans ,Immunology and Allergy ,Macrophage ,RNA, Messenger ,biology ,Macrophages ,Monocyte ,Brain ,Articles ,Transforming growth factor beta ,Blotting, Northern ,medicine.anatomical_structure ,Cytokine ,Astrocytes ,HIV-1 ,biology.protein ,Transforming growth factor ,Astrocyte - Abstract
The multifunctional cytokine, transforming growth factor beta (TGF-beta), was identified by immunocytochemistry in the brain tissues of four patients with acquired immune deficiency syndrome (AIDS), but not in control brain tissue. The TGF-beta staining was localized to cells of monocytic lineage as well as astrocytes, especially in areas of brain pathology. In addition, the brain tissues from the AIDS patients contained transcripts for human immunodeficiency virus 1 (HIV-1) by in situ hybridization, suggesting a correlation between the presence of HIV-1 in the brain and the expression of TGF-beta. However, the expression of TGF-beta was not limited to HIV-1-positive cells, raising the possibility of alternative mechanisms for the induction of TGF-beta in these AIDS patients' brains. To investigate these mechanisms, purified human monocytes were infected in vitro with HIV-1 and were shown to secrete increased levels of TGF-beta. In addition, HIV-1-infected monocytes released a factor(s) capable of triggering cultured astrocytes that are not infected with HIV-1 to secrete TGF-beta. The release of TGF-beta, which is an extremely potent chemotactic factor, may contribute to the recruitment of HIV-1-infected monocytic cells, enabling viral spread to and within the central nervous system (CNS). Moreover, TGF-beta augments cytokine production, including cytokines known to be neurotoxic. The identification of TGF-beta within the CNS implicates this cytokine in the immunopathologic processes responsible for AIDS-related CNS dysfunction.
- Published
- 1991
48. Activin a release into cerebrospinal fluid in a subset of patients with severe traumatic brain injury
- Author
-
Alexios A Adamides, Nicole Bye, Maria Cristina Morganti-Kossmann, Phuong Nguyen, David James Phillips, Jeffrey V. Rosenfeld, Lynnette Murray, Shirley Vallance, and Thomas Kossmann
- Subjects
Adult ,Glycerol ,Male ,endocrine system ,medicine.medical_specialty ,Follistatin ,Adolescent ,Traumatic brain injury ,Glutamic Acid ,Inflammation ,S100 Calcium Binding Protein beta Subunit ,Neuroprotection ,Cerebrospinal fluid ,Internal medicine ,Pyruvic Acid ,medicine ,Humans ,Lactic Acid ,Nerve Growth Factors ,Aged ,Trauma Severity Indices ,biology ,business.industry ,S100 Proteins ,Glasgow Coma Scale ,Middle Aged ,medicine.disease ,Activins ,Radiography ,Endocrinology ,Nerve growth factor ,Brain Injuries ,Case-Control Studies ,Phosphopyruvate Hydratase ,embryonic structures ,biology.protein ,Female ,Neurology (clinical) ,medicine.symptom ,business ,hormones, hormone substitutes, and hormone antagonists ,Transforming growth factor - Abstract
Activin A is a member of the transforming growth factor-beta superfamily and has been demonstrated to be elevated during inflammation and to have neuroprotective properties following neural insults. In this study, we examined whether traumatic brain injury (TBI) induced a response in activin A or in the concentrations of its binding protein, follistatin. Thirty-nine patients with severe TBI had daily, matched cerebrospinal fluid (CSF) and serum samples collected post-TBI and these were assayed for activin A and follistatin using specific immunoassays. Concentrations of both molecules were assessed relative to a variety of clinical parameters, such as the Glasgow Coma Score, computer tomography classification of TBI, measurement of injury markers, cell metabolism and membrane breakdown products. In about half of the patients, there was a notable increase in CSF activin A concentrations in the first few days post-TBI. There were only minor perturbations in either serum activin or in either CSF or serum follistatin concentrations. The CSF activin A response was not related to any of the common TBI indices, but was strongly correlated with two common markers of brain damage, neuronal specific enolase and S100-beta. Further, activin A levels were also associated with indices of metabolism, such as lactate and pyruvate, excitotoxicity (glutamate) and membrane lipid breakdown products such as glycerol. In one of the two patients who developed a CSF infection, activin A concentrations in CSF became markedly elevated. Thus, some TBI patients have an early release of activin A into the CSF that may result from activation of inflammatory and/or neuroprotective pathways.
- Published
- 2006
49. Detection of erythropoietin in human liquor: Intrinsic erythropoietin production in the brain
- Author
-
Max Gassmann, Otmar Trentz, Roland H. Wenger, Hugo H. Marti, Maria Cristina Morganti-Kossmann, Christian Bauer, Thomas Kossmann, Ivica Kvietikova, University of Zurich, and Marti, H H
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Central nervous system ,10052 Institute of Physiology ,Mice ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Cerebrospinal fluid ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Animals ,Craniocerebral Trauma ,Humans ,RNA, Messenger ,Receptor ,Erythropoietin ,030304 developmental biology ,0303 health sciences ,Kidney ,2727 Nephrology ,business.industry ,Brain ,Middle Aged ,3. Good health ,medicine.anatomical_structure ,Cytokine ,Endocrinology ,Blood-Brain Barrier ,Nephrology ,570 Life sciences ,biology ,Erythropoiesis ,Female ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Detection of erythropoietin in human liquor: Intrinsic erythropoietin production in the brain. Until now, erythropoietin (EPO) was thought to be produced exclusively in fetal liver and adult kidney and to regulate mammalian erythropoiesis. However, we recently showed that steady state levels of EPO mRNA could be induced up to 100-fold in primary mouse astrocytes cultured under hypoxic conditions, and also reported the presence of mRNA for EPO and its receptor in the brain of mouse, monkey and human. In extending these studies on humans we now show that immunoreactive EPO is present in ventricular cerebrospinal fluid (CSF) of 5 patients with traumatic brain injuries: EPO was found in 15 out of 15 CSF samples. There was no correlation between the serum EPO concentration and the concentration in the CSF. However, EPO concentrations in CSF correlated with the degree of blood-brain-barrier dysfunction. This suggests that EPO does not cross the intact blood-brain-barrier, implying that EPO is produced in the brain itself, most probably by astrocytes in an oxygen-dependent manner. In view that neuronal cells carry the EPO receptor, we propose that EPO acts in a paracrine fashion in the central nervous system and might function as a protective factor against hypoxia-induced damage of neurons.
- Published
- 1997
50. Influence of Brain Trauma on Blood—Brain Barrier Properties
- Author
-
Maria Cristina Morganti-Kossmann, Thomas Kossmann, Phuong Nguyen, and Nicole Bye
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,medicine ,Blood–brain barrier ,business ,Brain trauma - Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.