Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 34(11): 1791-801, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25160672

ABSTRACT

Focal cerebral ischemia is among the main causes of death and disability worldwide. The ischemic core often progresses, invading the peri-ischemic brain; however, assessing the propensity of the peri-ischemic brain to undergo secondary damage, understanding the underlying mechanisms, and adjusting treatment accordingly remain clinically unmet challenges. A significant hallmark of the peri-ischemic brain is dysfunction of the blood-brain barrier (BBB), yet the role of disturbed vascular permeability in stroke progression is unclear. Here we describe a longitudinal in vivo fluorescence imaging approach for the evaluation of cortical perfusion, BBB dysfunction, free radical formation and cellular injury using the photothrombosis vascular occlusion model in male Sprague Dawley rats. Blood-brain barrier dysfunction propagated within the peri-ischemic brain in the first hours after photothrombosis and was associated with free radical formation and cellular injury. Inhibiting free radical signaling significantly reduced progressive cellular damage after photothrombosis, with no significant effect on blood flow and BBB permeability. Our approach allows a dynamic follow-up of cellular events and their response to therapeutics in the acutely injured cerebral cortex.


Subject(s)
Blood-Brain Barrier , Brain Ischemia , Cerebral Cortex , Cerebrovascular Circulation , Intracranial Thrombosis , Stroke , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Free Radicals/metabolism , Intracranial Thrombosis/metabolism , Intracranial Thrombosis/pathology , Intracranial Thrombosis/physiopathology , Male , Permeability , Rats , Rats, Sprague-Dawley , Stroke/metabolism , Stroke/pathology , Stroke/physiopathology
2.
Article in English | MEDLINE | ID: mdl-24984896

ABSTRACT

Cell death is one of the pathophysiological hallmarks after stroke. Markers to image cell death pathways in vivo are highly desirable. We previously showed that fluorescently labeled Annexin A5 (AnxA5), which binds specifically to phosphatidylserine (PS) on dead/dying cells, can be used in experimental stroke for monitoring cell death with optical imaging. Here we investigated whether dual-labeled AnxA5 (technetium and fluorescence label) can be used for single-photon emission computed tomography (SPECT) of cell death in the same model. C57Bl6/N mice were subjected to 60-minute middle cerebral artery occlusion (MCAO) and underwent SPECT imaging at 24, 48, and 72 hours afterwards. They were injected intravenously with either PS-binding AnxA5 or the nonfunctional AnxA5 (negative control), labeled with 99mTc and Alexa Fluor 568, respectively. After SPECT imaging, brain sections were cut for autoradiography and fluorescence microscopy. Ethanol-induced cell death in the femur muscle was used as positive control. We detected dual-labeled AnxA5 in the model of ethanol-induced cell death in the femur muscle, but not after MCAO at any time point, either with SPECT or with ex vivo autoradiography or fluorescence microscopy. Dual-labeled AnxA5 appears to be unsuited for visualizing death of brain cells in this MCAO model.


Subject(s)
Annexin A5/pharmacology , Cerebral Angiography , Fluorescent Dyes/pharmacology , Stroke/diagnostic imaging , Technetium/pharmacology , Tomography, Emission-Computed, Single-Photon , Animals , Annexin A5/chemistry , Cell Death , Contrast Media/chemistry , Contrast Media/pharmacology , Disease Models, Animal , Fluorescent Dyes/chemistry , Male , Mice , Microscopy, Fluorescence , Stroke/metabolism , Technetium/chemistry , Time Factors
3.
J Cereb Blood Flow Metab ; 34(1): 144-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24129748

ABSTRACT

Neuronal damage shortly after onset or after brief episodes of cerebral ischemia has remained difficult to assess with clinical and preclinical imaging techniques as well as with microscopical methods. We here show, in rodent models of middle cerebral artery occlusion (MCAO), that neuronal damage in acute focal cerebral ischemia can be mapped with single-cell resolution using thallium autometallography (TlAMG), a histochemical technique for the detection of the K(+)-probe thallium (Tl(+)) in the brain. We intravenously injected rats and mice with thallium diethyldithiocarbamate (TlDDC), a lipophilic chelate complex that releases Tl(+) after crossing the blood-brain barrier. We found, within the territories of the affected arteries, areas of markedly reduced neuronal Tl(+) uptake in all animals at all time points studied ranging from 15 minutes to 24 hours after MCAO. In large lesions at early time points, areas with neuronal and astrocytic Tl(+) uptake below thresholds of detection were surrounded by putative penumbral zones with preserved but diminished Tl(+) uptake. At 24 hours, the areas of reduced Tl(+)uptake matched with areas delineated by established markers of neuronal damage. The results suggest the use of (201)TlDDC for preclinical and clinical single-photon emission computed tomography (SPECT) imaging of hyperacute alterations in brain K(+) metabolism and prediction of tissue viability in cerebral ischemia.


Subject(s)
Brain/pathology , Chelating Agents , Ditiocarb , Ischemic Attack, Transient/pathology , Neurons/pathology , Thallium , Acute Disease , Animals , Autoradiography , Brain/diagnostic imaging , Brain/metabolism , Brain Mapping , Chelating Agents/administration & dosage , Disease Models, Animal , Ditiocarb/administration & dosage , Histocytochemistry/methods , Ischemic Attack, Transient/diagnostic imaging , Ischemic Attack, Transient/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/diagnostic imaging , Neurons/metabolism , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Thallium Radioisotopes/administration & dosage , Tomography, Emission-Computed, Single-Photon/methods
4.
PLoS One ; 8(8): e69529, 2013.
Article in English | MEDLINE | ID: mdl-23936336

ABSTRACT

Inflammation is a pathophysiological hallmark of many diseases of the brain. Specific imaging of cells and molecules that contribute to cerebral inflammation is therefore highly desirable, both for research and in clinical application. The 18 kDa translocator protein (TSPO) has been established as a suitable target for the detection of activated microglia/macrophages. A number of novel TSPO ligands have been developed recently. Here, we evaluated the high affinity TSPO ligand DPA-714 as a marker of brain inflammation in two independent animal models. For the first time, the specificity of radiolabeled DPA-714 for activated microglia/macrophages was studied in a rat model of epilepsy (induced using Kainic acid) and in a mouse model of stroke (transient middle cerebral artery occlusion, tMCAO) using high-resolution autoradiography and immunohistochemistry. Additionally, cold-compound blocking experiments were performed and changes in blood-brain barrier (BBB) permeability were determined. Target-to-background ratios of 2 and 3 were achieved in lesioned vs. unaffected brain tissue in the epilepsy and tMCAO models, respectively. In both models, ligand uptake into the lesion corresponded well with the extent of Ox42- or Iba1-immunoreactive activated microglia/macrophages. In the epilepsy model, ligand uptake was almost completely blocked by pre-injection of DPA-714 and FEDAA1106, another high-affinity TSPO ligand. Ligand uptake was independent of the degree of BBB opening and lesion size in the stroke model. We provide further strong evidence that DPA-714 is a specific ligand to image activated microglia/macrophages in experimental models of brain inflammation.


Subject(s)
Carrier Proteins/metabolism , Disease Models, Animal , Epilepsy/complications , Inflammation/diagnosis , Pyrazoles , Pyrimidines , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Stroke/complications , Animals , Autoradiography , Blood-Brain Barrier , Cells, Cultured , Epilepsy/chemically induced , Epilepsy/pathology , Fluorine Radioisotopes , Immunoenzyme Techniques , Inflammation/etiology , Inflammation/metabolism , Kainic Acid/toxicity , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Positron-Emission Tomography , Pyrazoles/metabolism , Pyrimidines/metabolism , Radiopharmaceuticals , Rats , Rats, Sprague-Dawley , Stroke/chemically induced , Stroke/pathology
5.
J Cereb Blood Flow Metab ; 31(5): 1311-20, 2011 May.
Article in English | MEDLINE | ID: mdl-21245871

ABSTRACT

To monitor stroke-induced brain damage and assess neuroprotective therapies, specific imaging of cell death after cerebral ischemia in a noninvasive manner is highly desirable. Annexin A5 has been suggested as a marker for imaging cell death under various disease conditions including stroke. In this study, C57BL6/N mice received middle cerebral artery occlusion (MCAO) and were injected intravenously with either active or inactive Cy5.5-annexin A5 48 hours after reperfusion. Some mice also received propidium iodide (PI), a cell integrity marker. Only in mice receiving active Cy5.5-annexin A5 were fluorescence intensities significantly higher over the hemisphere ipsilateral to MCAO than on the contralateral side. This was detected noninvasively and ex vivo 4 and 8 hours after injection. The majority of cells positive for fluorescent annexin A5 were also positive for PI and fragmented DNA as detected by terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL) staining. This study demonstrates the high specificity of annexin A5 for visualization of cell death in a mouse model of stroke. To our knowledge, this is the first study to compare the distribution of injected active and inactive annexin A5, PI, and TUNEL staining. It provides important information on the experimental and potential clinical applications of annexin A5-based imaging agents in stroke.


Subject(s)
Annexin A5 , Brain Ischemia/pathology , Fluorescent Dyes , Microscopy, Fluorescence/methods , Propidium , Animals , Biomarkers/analysis , Carbocyanines , Cell Death/physiology , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Staining and Labeling/methods
6.
J Cereb Blood Flow Metab ; 31(2): 757-66, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20842165

ABSTRACT

Reduced infarct volume in TLR2-knockout mice compared with C57Bl/6 wild-type mice has recently been shown in experimental stroke and confirmed in this study. We now also show a significant decrease of CD11b-positive cell counts and decreased neuronal death in the ischemic hemispheres of TLR2-deficient mice compared with C57Bl/6wt mice 2 days after transient focal cerebral ischemia. To examine the potential benefit of intravascular TLR2 inhibition, C57Bl/6wt mice were treated intraarterially with TLR2-blocking anti-TLR2 antibody (clone T2.5) after 45 minutes of cerebral ischemia and compared with control antibody (isotype) treated wild-type mice. Whereas T2.5-treated mice had no reduction in infarct volumes at 48 hours after reperfusion, they did have decreased numbers of CD11b-positive inflammatory cells and decreased neuronal death compared with isotype-treated control mice. Comparison of the isotype antibody treatment to control (saline) treatment showed no effects on infarct volumes or neuronal survival. However, mice treated with the control isotype antibody had increased numbers of CD11b-positive inflammatory cells compared with saline-treated animals. Thus, antibody treatment itself (i.e., control isotype antibody, but potentially of any antibody) may have adverse effects and limit therapeutic benefit of anti-TLR2-antibody therapy. We conclude that TLR2 mediates leukocyte and microglial infiltration and neuronal death, which can be attenuated by TLR2 inhibition. The TLR2 inhibition in vivo improves neuronal survival and may represent a future stroke therapy.


Subject(s)
Inflammation/pathology , Neurons/pathology , Stroke/drug therapy , Stroke/pathology , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/physiology , Animals , Antibodies, Blocking/pharmacology , Brain Edema/pathology , Brain Edema/prevention & control , CD11b Antigen/metabolism , Cell Count , Cell Death/drug effects , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 2/genetics
7.
J Immunol ; 184(11): 6407-17, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20439917

ABSTRACT

The stem-cell marker CD93 (AA4.1/C1qRp) has been described as a potential complement C1q-receptor. Its exact molecular function, however, remains unknown. By using global expression profiling we showed that CD93-mRNA is highly induced after transient focal cerebral ischemia. CD93 protein is upregulated in endothelial cells, but also in selected macrophages and microglia. To elucidate the potential functional role of CD93 in postischemic brain damage, we used mice with a targeted deletion of the CD93 gene. After 30 min of occlusion of the middle cerebral artery and 3 d of reperfusion these mice displayed increased leukocyte infiltration into the brain, increased edema, and significantly larger infarct volumes (60.8 +/- 52.2 versus 23.9 +/- 16.6 mm(3)) when compared with wild-type (WT) mice. When the MCA was occluded for 60 min, after 2 d of reperfusion the CD93 knockout mice still showed more leukocytes in the brain, but the infarct volumes were not different from those seen in WT animals. To further explore CD93-dependent signaling pathways, we determined global transcription profiles and compared CD93-deficient and WT mice at various time points after induction of focal cerebral ischemia. We found a highly significant upregulation of the chemokine CCL21/Exodus-2 in untreated and treated CD93-deficient mice at all time points. Induction of CCL21 mRNA and protein was confirmed by PCR and immunohistochemistry. CCL21, which was formerly shown to be released by damaged neurons and to activate microglia, contributes to neurodegeneration. Thus, we speculate that CD93-neuroprotection is mediated via suppression of the neuroinflammatory response through downregulation of CCL21.


Subject(s)
Brain Ischemia/genetics , Inflammation/genetics , Membrane Glycoproteins/biosynthesis , Receptors, Complement/biosynthesis , Animals , Blotting, Western , Brain Ischemia/metabolism , Brain Ischemia/pathology , Chemokine CCL21/biosynthesis , Chemokine CCL21/genetics , Chemokine CCL21/immunology , Female , Gene Expression , Gene Expression Profiling , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Receptors, Complement/genetics , Receptors, Complement/immunology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reverse Transcriptase Polymerase Chain Reaction
8.
J Neuroinflammation ; 7: 15, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20202211

ABSTRACT

BACKGROUND: The complement system is a crucial mediator of inflammation and cell lysis after cerebral ischemia. However, there is little information about the exact contribution of the membrane attack complex (MAC) and its inhibitor-protein CD59. METHODS: Transient focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in young male and female CD59a knockout and wild-type mice. Two models of MCAO were applied: 60 min MCAO and 48 h reperfusion, as well as 30 min MCAO and 72 h reperfusion. CD59a knockout animals were compared to wild-type animals in terms of infarct size, edema, neurological deficit, and cell death. RESULTS AND DISCUSSION: CD59a-deficiency in male mice caused significantly increased infarct volumes and brain swelling when compared to wild-type mice at 72 h after 30 min-occlusion time, whereas no significant difference was observed after 1 h-MCAO. Moreover, CD59a-deficient mice had impaired neurological function when compared to wild-type mice after 30 min MCAO. CONCLUSION: We conclude that CD59a protects against ischemic brain damage, but depending on the gender and the stroke model used.


Subject(s)
Brain Infarction/metabolism , CD59 Antigens/metabolism , Infarction, Middle Cerebral Artery/metabolism , Animals , Brain Infarction/etiology , Brain Infarction/pathology , CD11b Antigen/metabolism , CD59 Antigens/genetics , Disease Models, Animal , Female , In Situ Nick-End Labeling/methods , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurologic Examination/methods , Sex Factors , Time Factors
9.
Biomed Opt Express ; 1(1): 97-105, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-21258449

ABSTRACT

Non-invasive near-infrared fluorescence (NIRF) imaging is a powerful tool to study pathophysiology in a wide variety of animal disease models including brain diseases. However, especially in NIRF imaging of the brain or other deeper laying target sites, background fluorescence emitted from the scalp or superficial blood vessels can impede the detection of fluorescence in deeper tissue. Here, we introduce an effective method to reduce the impact of fluorescence from superficial layers. The approach uses excitation light at two different wavelengths generating two images with different depth sensitivities followed by an adapted subtraction algorithm. This technique leads to significant enhancement of the contrast and the detectability of fluorochromes located in deep tissue layers in tissue simulating phantoms and murine models with stroke.

10.
Biochim Biophys Acta ; 1792(12): 1198-204, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19835955

ABSTRACT

Several reports have recently demonstrated a detrimental role of Toll-like receptors (TLR) in cerebral ischemia, while there is little information about the endogenous ligands which activate TLR-signaling. The myeloid related proteins-8 and-14 (Mrp8/S100A8; Mrp14/S100A9) have recently been characterized as endogenous TLR4-agonists, and thus may mediate TLR-activation in cerebral ischemia. Interestingly, not only TLR-mRNAs, but also Mrp8 and Mrp14 mRNA were found to be induced in mouse brain between 3 and 48 h after transient 1 h focal cerebral ischemia/reperfusion. Mrp-protein was expressed in the ischemic hemisphere, and co-labeled with CD11b-positive cells. To test the hypothesis that Mrp-signaling contributes to the postischemic brain damage, we subjected Mrp14-deficient mice, which also lack Mrp8 protein expression, to focal cerebral ischemia. Mrp14-deficient mice had significantly smaller lesion volumes when compared to wild-type littermates (130+/-16 mm(3) vs. 105+/-28 mm(3)) at 2 days after transient focal cerebral ischemia (1 h), less brain swelling, and a reduced macrophage/microglia cell count in the ischemic hemisphere. We conclude that upregulation and signaling of Mrp-8 and-14 contribute to neuroinflammation and the progression of ischemic damage.


Subject(s)
Brain Ischemia/metabolism , Calgranulin A/physiology , Calgranulin B/physiology , Central Nervous System/injuries , Inflammation/etiology , Neurons/pathology , Animals , Brain Ischemia/pathology , Central Nervous System/metabolism , Gene Expression Profiling , Immunoenzyme Techniques , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Biochem Biophys Res Commun ; 359(3): 574-9, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17548055

ABSTRACT

A significant up-regulation of Toll-like-receptor (TLR) mRNAs between 3 and 48 h reperfusion time after induction of transient focal cerebral ischemia for 1h was revealed by applying global gene expression profiling in postischemic mouse brains. Compared to TLR4 and TLR9, TLR2 proved to be the most significantly up-regulated TLR in the ipsilateral brain hemisphere. TLR2-protein was found to be expressed mainly in microglia in the postischemic brain tissue, but also in selected endothelial cells, neurons, and astrocytes. Additionally, TLR2-related genes with pro-inflammatory and pro-apoptotic capabilities were induced. Therefore we hypothesized that TLR2-signaling could exacerbate the primary brain damage after ischemia. Two days after induction of transient focal cerebral ischemia (1h), we found a significant decrease of the infarct volume in TLR2 deficient mice compared to wild type mice (75+/-5 vs. 42+/-7 mm(3)). We conclude that TLR2 up-regulation and TLR2-signaling are important events in focal cerebral ischemia and contribute to the deterioration of ischemic damage.


Subject(s)
Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Toll-Like Receptor 2/metabolism , Animals , Ischemic Attack, Transient/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Up-Regulation
12.
J Neuroinflammation ; 4: 13, 2007 May 02.
Article in English | MEDLINE | ID: mdl-17474994

ABSTRACT

BACKGROUND: The posttraumatic response to traumatic brain injury (TBI) is characterized, in part, by activation of the innate immune response, including the complement system. We have recently shown that mice devoid of a functional alternative pathway of complement activation (factor B-/- mice) are protected from complement-mediated neuroinflammation and neuropathology after TBI. In the present study, we extrapolated this knowledge from studies in genetically engineered mice to a pharmacological approach using a monoclonal anti-factor B antibody. This neutralizing antibody represents a specific and potent inhibitor of the alternative complement pathway in mice. METHODS: A focal trauma was applied to the left hemisphere of C57BL/6 mice (n = 89) using a standardized electric weight-drop model. Animals were randomly assigned to two treatment groups: (1) Systemic injection of 1 mg monoclonal anti-factor B antibody (mAb 1379) in 400 mul phosphate-buffered saline (PBS) at 1 hour and 24 hours after trauma; (2) Systemic injection of vehicle only (400 mul PBS), as placebo control, at identical time-points after trauma. Sham-operated and untreated mice served as additional negative controls. Evaluation of neurological scores and analysis of brain tissue specimens and serum samples was performed at defined time-points for up to 1 week. Complement activation in serum was assessed by zymosan assay and by murine C5a ELISA. Brain samples were analyzed by immunohistochemistry, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) histochemistry, and real-time RT-PCR. RESULTS: The mAb 1379 leads to a significant inhibition of alternative pathway complement activity and to significantly attenuated C5a levels in serum, as compared to head-injured placebo-treated control mice. TBI induced histomorphological signs of neuroinflammation and neuronal apoptosis in the injured brain hemisphere of placebo-treated control mice for up to 7 days. In contrast, the systemic administration of an inhibitory anti-factor B antibody led to a substantial attenuation of cerebral tissue damage and neuronal cell death. In addition, the posttraumatic administration of the mAb 1379 induced a neuroprotective pattern of intracerebral gene expression. CONCLUSION: Inhibition of the alternative complement pathway by posttraumatic administration of a neutralizing anti-factor B antibody appears to represent a new promising avenue for pharmacological attenuation of the complement-mediated neuroinflammatory response after head injury.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Injuries/immunology , Brain Injuries/prevention & control , Complement Factor B/antagonists & inhibitors , Complement Factor B/immunology , Complement Pathway, Alternative/immunology , Animals , Male , Mice , Mice, Inbred C57BL
13.
BMC Neurosci ; 7: 55, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16842619

ABSTRACT

BACKGROUND: Neuroprotective strategies for prevention of the neuropathological sequelae of traumatic brain injury (TBI) have largely failed in translation to clinical treatment. Thus, there is a substantial need for further understanding the molecular mechanisms and pathways which lead to secondary neuronal cell death in the injured brain. The intracerebral activation of the complement cascade was shown to mediate inflammation and tissue destruction after TBI. However, the exact pathways of complement activation involved in the induction of posttraumatic neurodegeneration have not yet been assessed. In the present study, we investigated the role of the alternative complement activation pathway in contributing to neuronal cell death, based on a standardized TBI model in mice with targeted deletion of the factor B gene (fB-/-), a "key" component required for activation of the alternative complement pathway. RESULTS: After experimental TBI in wild-type (fB+/+) mice, there was a massive time-dependent systemic complement activation, as determined by enhanced C5a serum levels for up to 7 days. In contrast, the extent of systemic complement activation was significantly attenuated in fB-/- mice (P < 0.05,fB-/- vs. fB+/+; t = 4 h, 24 h, and 7 days after TBI). TUNEL histochemistry experiments revealed that posttraumatic neuronal cell death was clearly reduced for up to 7 days in the injured brain hemispheres of fB-/- mice, compared to fB+/+ littermates. Furthermore, a strong upregulation of the anti-apoptotic mediator Bcl-2 and downregulation of the pro-apoptotic Fas receptor was detected in brain homogenates of head-injured fB-/- vs. fB+/+ mice by Western blot analysis. CONCLUSION: The alternative pathway of complement activation appears to play a more crucial role in the pathophysiology of TBI than previously appreciated. This notion is based on the findings of (a) the significant attenuation of overall complement activation in head-injured fB-/- mice, as determined by a reduction of serum C5a concentrations to constitutive levels in normal mice, and (b) by a dramatic reduction of TUNEL-positive neurons in conjunction with an upregulation of Bcl-2 and downregulation of the Fas receptor in head-injured fB-/- mice, compared to fB+/+ littermates. Pharmacological targeting of the alternative complement pathway during the "time-window of opportunity" after TBI may represent a promising new strategy to be pursued in future studies.


Subject(s)
Brain Injuries/physiopathology , Brain/physiopathology , Complement Factor B/metabolism , Neurons , Animals , Brain/metabolism , Brain Injuries/metabolism , Cell Death , Complement Activation , Complement Factor B/deficiency , Down-Regulation , Head Injuries, Closed/metabolism , Head Injuries, Closed/physiopathology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...