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










Publication year range
2.
J Exp Med ; 210(1): 157-72, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23296467

ABSTRACT

Colony-stimulating factor 1 (CSF1) and interleukin-34 (IL-34) are functional ligands of the CSF1 receptor (CSF1R) and thus are key regulators of the monocyte/macrophage lineage. We discovered that systemic administration of human recombinant CSF1 ameliorates memory deficits in a transgenic mouse model of Alzheimer's disease. CSF1 and IL-34 strongly reduced excitotoxin-induced neuronal cell loss and gliosis in wild-type mice when administered systemically before or up to 6 h after injury. These effects were accompanied by maintenance of cAMP responsive element-binding protein (CREB) signaling in neurons rather than in microglia. Using lineage-tracing experiments, we discovered that a small number of neurons in the hippocampus and cortex express CSF1R under physiological conditions and that kainic acid-induced excitotoxic injury results in a profound increase in neuronal receptor expression. Selective deletion of CSF1R in forebrain neurons in mice exacerbated excitotoxin-induced death and neurodegeneration. We conclude that CSF1 and IL-34 provide powerful neuroprotective and survival signals in brain injury and neurodegeneration involving CSF1R expression on neurons.


Subject(s)
Macrophage Colony-Stimulating Factor/pharmacology , Neurons/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Base Sequence , Cell Survival , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Humans , Interleukins/genetics , Interleukins/pharmacology , Kainic Acid/toxicity , Macrophage Colony-Stimulating Factor/administration & dosage , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphorylation , Prosencephalon/metabolism , Prosencephalon/pathology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction
3.
Biotechnol Appl Biochem ; 57(1): 31-45, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20815818

ABSTRACT

NgRI (Nogo-66 receptor) is part of a signalling complex that inhibits axon regeneration in the central nervous system. Truncated soluble versions of NgRI have been used successfully to promote axon regeneration in animal models of spinal-cord injury, raising interest in this protein as a potential therapeutic target. The LRR (leucine-rich repeat) regions in NgRI are flanked by N- and C-terminal disulfide-containing 'cap' domains (LRRNT and LRRCT respectively). In the present work we show that, although functionally active, the NgRI(310)-Fc fusion protein contains mislinked and heterogeneous disulfide patterns in the LRRCT domain, and we report the generation of a series of variant molecules specifically designed to prevent this heterogeneity. Using these variants we explored the effects of modifying the NgRI truncation site or the spacing between the NgRI and Fc domains, or replacing cysteines within the NgRI or IgG hinge regions. One variant, which incorporates replacements of Cys²66 and Cys³°9 with alanine residues, completely eliminated disulfide scrambling while maintaining functional in vitro and in vivo efficacy. This modified NgRI-Fc molecule represents a significantly improved candidate for further pharmaceutical development, and may serve as a useful model for the optimization of other IgG fusion proteins made from LRR proteins.


Subject(s)
Disulfides/metabolism , Myelin Proteins/chemistry , Protein Engineering/methods , Receptors, Cell Surface/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Behavior, Animal/drug effects , Crystallography, X-Ray , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , Humans , Male , Molecular Sequence Annotation , Molecular Sequence Data , Myelin Proteins/genetics , Nogo Receptor 1 , Protein Stability , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Nerve Roots/injuries
4.
Brain Behav Immun ; 24(5): 738-46, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20138139

ABSTRACT

Non-CNS chemokine production may contribute to previously unrecognised components of Multiple Sclerosis (MS) pathology. Here we show that IL-8, a neutrophil chemoattractant, is significantly increased in serum from individuals with MS, and that the rodent homolog of IL-8 (CXCL1) is expressed in the liver in experimental autoimmune encephalomyelitis (EAE), a rodent model of MS. The hepatic expression of CXCL1 in EAE is accompanied by neutrophil recruitment to the liver, and we show that this recruitment is a feature of post mortem liver tissue from MS patients, which is a previously unrecognised phenomenon. We speculated that the presence of peripheral CXC-chemokine expression might contribute to the sickness behaviours associated with MS, which are a significant contributor to morbidity. Peripheral, but not central, administration of CXCL1 to Wistar rats inhibited spontaneous activity in the open field and burrowing behaviour in a dose-dependent manner (5-45 microg). The expression of CXCL1 by the liver and the recruitment of neutrophils can be modelled by the intracerebral injection of IL-1beta. Here, we found that interferon-beta (IFN-beta) pretreatment significantly inhibited hepatic CXCL1 production and neutrophil recruitment to the liver induced by the microinjection of IL-1beta into the brain. Thus while the mechanism by which IFN-beta therapy suppresses disease in MS remains unclear, the data presented here suggests that the inhibition of hepatic chemokine synthesis may be a contributing factor.


Subject(s)
Chemokine CXCL1/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Illness Behavior , Interleukin-8/blood , Liver/metabolism , Multiple Sclerosis/blood , Analysis of Variance , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/immunology , Brain/metabolism , Chemokine CXCL1/immunology , Chemokine CXCL1/pharmacology , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Immunohistochemistry , Immunologic Factors/pharmacology , Interferon-beta/pharmacology , Interleukin-1beta/pharmacology , Interleukin-8/immunology , Liver/immunology , Male , Motor Activity/drug effects , Multiple Sclerosis/immunology , Neutrophils/immunology , Neutrophils/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
5.
J Cereb Blood Flow Metab ; 30(1): 130-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19773800

ABSTRACT

Inflammatory mechanisms are known to contribute to the pathophysiology of traumatic brain injury (TBI). Since bradykinin is one of the first mediators activated during inflammation, we investigated the role of bradykinin and its receptors in posttraumatic secondary brain damage. We subjected wild-type (WT), B(1)-, and B(2)-receptor-knockout mice to controlled cortical impact (CCI) and analyzed tissue bradykinin as well as kinin receptor mRNA and protein expression up to 48 h thereafter. Brain edema, contusion volume, and functional outcome were assessed 24 h and 7 days after CCI. Tissue bradykinin was maximally increased 2 h after trauma (P<0.01 versus sham). Kinin B(1) receptor mRNA was upregulated up to four-fold 24 h after CCI. Immunohistochemistry showed that B(1) and B(2) receptors were expressed in the brain and were significantly upregulated in the traumatic penumbra 1 to 24 h after CCI. B(2)R(-/-) mice had significantly less brain edema (-51% versus WT, 24 h; P<0.001), smaller contusion volumes ( approximately 50% versus WT 24 h and 7 d after CCI; P<0.05), and better functional outcome 7 days after TBI as compared with WT mice (P<0.05). The present results show that bradykinin and its B(2) receptors play a causal role for brain edema formation and cell death after TBI.


Subject(s)
Brain Injuries/pathology , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Animals , Bradykinin/metabolism , Brain Edema/pathology , Contusions/pathology , Immunohistochemistry , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Bradykinin B1/biosynthesis , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/biosynthesis , Receptor, Bradykinin B2/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Brain ; 132(Pt 4): 1078-92, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19151372

ABSTRACT

Alzheimer's disease is a major cause of dementia in humans. The appearance of cognitive decline is linked to the overproduction of a short peptide called beta-amyloid (Abeta) in both soluble and aggregate forms. Here, we show that injecting macrophage colony-stimulating factor (M-CSF) to Swedish beta-amyloid precursor protein (APP(Swe))/PS1 transgenic mice, a well-documented model for Alzheimer's disease, on a weekly basis prior to the appearance of learning and memory deficits prevented cognitive loss. M-CSF also increased the number of microglia in the parenchyma and decreased the number of Abeta deposits. Senile plaques were smaller and less dense in the brain of M-CSF-treated mice compared to littermate controls treated with vehicle solution. Interestingly, a higher ratio of microglia internalized Abeta in the brain of M-CSF-treated animals and the phagocytosed peptides were located in the late endosomes and lysosomes. Less Abeta(40) and Abeta(42) monomers were also detected in the extracellular protein enriched fractions of M-CSF-treated transgenic mice when compared with vehicle controls. Finally, treating APP(Swe)/PS1 mice that were already demonstrating installed Abeta pathology stabilized the cognitive decline. Together these results provide compelling evidence that systemic M-CSF administration is a powerful treatment to stimulate bone marrow-derived microglia, degrade Abeta and prevent or improve the cognitive decline associated with Abeta burden in a mouse model of Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Cognition Disorders/prevention & control , Macrophage Colony-Stimulating Factor/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Behavior, Animal/drug effects , Brain/metabolism , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical/methods , Endosomes/metabolism , Male , Mice , Mice, Transgenic , Microglia/pathology
7.
Exp Neurol ; 214(2): 147-59, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19038604

ABSTRACT

The extent of disability caused by spinal cord injury (SCI) relates to secondary tissue destruction arising partly from an intraspinal influx of neutrophils and monocyte/macrophages after the initial injury. The integrin alpha4beta1, expressed by these leukocytes, is a key to their activation and migration into/within tissue. Therefore, blocking this integrin's functions may afford significant neuroprotection. Rats were treated intravenously with a blocking monoclonal antibody (mAb) to the alpha4 subunit of alpha4beta1 at 2 and 24 h after thoracic clip-compression SCI. Anti-alpha4beta1 treatment significantly decreased neutrophil and monocyte/macrophage influx at 3 d by 47% and 53%, respectively, and decreased neutrophil influx by 61% at 7 d after SCI. Anti-alpha4beta1 treatment also significantly reduced oxidative activity in injured cord homogenates at 3 d. For example, myeloperoxidase activity decreased by 38%, inducible nitric oxide by 44%, dichlorofluorescein (marking free radicals) by 33% and lipid peroxidation (malondialdehyde) by 42%. At 2-8 weeks after SCI, motor function improved by up to 2 points on an open-field locomotor scale. Treated rats supported weight with their hind paws instead of sweeping. At 2-4 weeks after SCI, anti-alpha4beta1 treatment decreased blood pressure responses during autonomic dysreflexia by as much as 43% and, at 2-8 weeks, decreased mechanical allodynia elicited from the trunk and hind paw by up to 54% and 40%, respectively. This improved functional recovery correlated with spared myelin-containing white matter and >10-fold more bulbospinal serotonergic axons below the injury than were in controls. The significant neurological improvement offered by this neuroprotective strategy underscores the potential for an anti-integrin treatment for SCI.


Subject(s)
Antibodies, Monoclonal/pharmacology , Integrin alpha4beta1/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/immunology , Spinal Cord/immunology , Acute Disease , Animals , Axons/pathology , Axons/physiology , Cell Count , Cell Movement/immunology , Female , Integrin alpha4beta1/immunology , Integrin alpha4beta1/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Monocytes/immunology , Monocytes/metabolism , Motor Activity , Neuralgia/drug therapy , Neuralgia/immunology , Neuralgia/pathology , Neutrophils/immunology , Neutrophils/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/immunology , Serotonin/physiology , Spinal Cord/pathology , Spinal Cord Injuries/pathology
8.
Mol Cell Neurosci ; 39(2): 258-67, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18692574

ABSTRACT

Oligodendrocyte-myelin glycoprotein (OMgp) is a myelin component that has been shown in vitro to inhibit neurite outgrowth by binding to the Nogo-66 receptor (NgR1)/Lingo-1/Taj (TROY)/p75 receptor complex to activate the RhoA pathway. To investigate the effects of OMgp on axon regeneration in vivo, OMgp(-/-) mice on a mixed 129/Sv/C57BL/6 (129BL6) or a C57BL/6 (BL6) genetic background were tested in two spinal cord injury (SCI) models - a severe complete transection or a milder dorsal hemisection. OMgp(-/-) mice on the mixed 129BL6 genetic background showed greater functional improvement compared to OMgp(+/+) littermates, with increased numbers of cholera toxin B-labeled ascending sensory axons and 5-HT(+) descending axons and less RhoA activation after spinal cord injury. Myelin isolated from OMgp(-/-) mice (129BL6) was significantly less inhibitory to neurite outgrowth than wild-type (wt) myelin in vitro. However, OMgp(-/-) mice on a BL/6 genetic background showed neither statistically significant functional recovery nor axonal sprouting following dorsal hemisection.


Subject(s)
Axons/physiology , Myelin-Associated Glycoprotein/deficiency , Recovery of Function/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Analysis of Variance , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Cholera Toxin/metabolism , Dextrans/metabolism , Exploratory Behavior/physiology , Female , Functional Laterality/genetics , GPI-Linked Proteins , Ganglia, Spinal/pathology , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Neurites/physiology , Neurons/pathology , Recovery of Function/genetics , Serotonin/metabolism , Time Factors , rhoA GTP-Binding Protein/metabolism
9.
Exp Gerontol ; 42(8): 762-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17592750

ABSTRACT

Neuroinflammation may play a role in the pathogenesis of Parkinson's disease (PD). The present study questioned whether this neuroinflammatory response differs between the olfactory bulb, as an early affected region and the nigrostriatal system. Indeed, increased microgliosis was shown in post-mortem olfactory bulb of PD patients. Also in olfactory bulb of MPTP-treated mice, microgliosis and increased expression of IL-1alpha, IL-1beta and IL-1ra mRNA was observed early after treatment. These observations implicate that neuroinflammation is not restricted to the nigrostriatal system. MPTP-induced microgliosis in striatum and olfactory bulb was reduced in IL-1alpha/beta knockout mice, indicating that IL-1 affects microglia activation. Importantly, MPTP induced differential regulation of IL-1 receptors. mRNA levels of IL-1RI and, to a lesser extent, IL-1RII were increased in striatum. Interestingly, in the olfactory bulb only IL-1RII mRNA was enhanced. We suggest that differential regulation of IL-1 signaling can serve as an important mechanism to modulate neuroinflammatory activity after MPTP treatment and possibly during PD.


Subject(s)
MPTP Poisoning/immunology , MPTP Poisoning/pathology , Olfactory Bulb/immunology , Olfactory Bulb/pathology , Parkinson Disease/immunology , Parkinson Disease/pathology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Receptors, Interleukin-1/genetics , Animals , Base Sequence , Corpus Striatum/drug effects , Corpus Striatum/immunology , Corpus Striatum/pathology , DNA Primers/genetics , Gene Expression , Humans , MPTP Poisoning/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Olfactory Bulb/drug effects , Parkinson Disease/genetics , Parkinsonian Disorders/genetics , Receptors, Interleukin-1/classification , Substantia Nigra/drug effects , Substantia Nigra/immunology , Substantia Nigra/pathology
10.
Mol Cell Neurosci ; 34(2): 147-54, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17150373

ABSTRACT

Integrins mediate cell adhesion to the extracellular matrix and initiate intracellular signaling. They play key roles in the central nervous system (CNS), participating in synaptogenesis, synaptic transmission and memory formation, but their precise mechanism of action remains unknown. Here we show that the integrin ligand-mimetic peptide GRGDSP induced NMDA receptor-dependent increases in intracellular calcium levels within seconds of presentation to primary cortical neurons. These were followed by transient activation and nuclear translocation of the ERK1/2 mitogen-activated protein kinase. RGD-induced effects were reduced by the NMDA receptor antagonist MK801, and ERK1/2 signaling was specifically inhibited by ifenprodil and PP2, indicating a functional connection between integrins, Src and NR2B-containing NMDA receptors. GRGDSP peptides were not significantly neuroprotective against excitotoxic insults. These results demonstrate a previously undescribed, extremely rapid effect of RGD peptide binding to integrins on cortical neurons that implies a close, functionally relevant connection between adhesion receptors and synaptic transmission.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium/metabolism , Cerebral Cortex/cytology , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/drug effects , Oligopeptides/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Animals , Cell Death/drug effects , Cells, Cultured , Embryo, Mammalian , Hydro-Lyases/metabolism , Immunohistochemistry , Rats
11.
Mol Cell Neurosci ; 33(3): 311-20, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17011208

ABSTRACT

LINGO-1 is a CNS-specific protein and a functional component of the NgR1/p75/LINGO-1 and NgR1/TAJ(TROY)/LINGO-1 signaling complexes that mediate inhibition of axonal outgrowth. These receptor complexes mediate the axonal growth inhibitory effects of Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte-myelin glycoprotein (OMgp) via RhoA activation. Soluble LINGO-1 (LINGO-1-Fc), which acts as an antagonist of these pathways by blocking LINGO-1 binding to NgR1, was administered to rats after dorsal or lateral hemisection of the spinal cord. LINGO-1-Fc treatment significantly improved functional recovery, promoted axonal sprouting and decreased RhoA activation and increased oligodendrocyte and neuronal survival after either rubrospinal or corticospinal tract transection. These experiments demonstrate an important role for LINGO-1 in modulating axonal outgrowth in vivo and that treatment with LINGO-1-Fc can significantly enhance recovery after spinal cord injury.


Subject(s)
Axons/drug effects , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Analysis of Variance , Animals , Apoptosis/drug effects , Axons/physiology , Caspase 3/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Forelimb/drug effects , Humans , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , MAP Kinase Kinase 4/metabolism , Membrane Proteins/chemistry , Membrane Proteins/physiology , Nerve Regeneration/drug effects , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Organogenesis/drug effects , Protein Binding/drug effects , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Time Factors , Tubulin/metabolism , rhoA GTP-Binding Protein/metabolism
12.
Eur J Neurosci ; 22(3): 587-94, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16101740

ABSTRACT

Methylprednisolone (MP) is a synthetic glucocorticoid used for the treatment of spinal cord injury (SCI). Soluble Nogo-66 receptor (NgR) ectodomain is a novel experimental therapy for SCI that promotes axonal regeneration by blocking the growth inhibitory effects of myelin constituents in the adult central nervous system. To evaluate the potential complementarity of these mechanistically distinct pharmacological reagents we compared their effects alone and in combination after thoracic (T7) dorsal hemisection in the rat. Treatment with an ecto-domain of the rat NgR (27-310) fused to a rat IgG [NgR(310)ecto-Fc] (50 microm intrathecal, 0.25 microL/h for 28 days) or MP alone (30 mg/kg i.v., 0, 4 and 8 h postinjury) improved the rate and extent of functional recovery measured using Basso, Beattie, Bresnahan (BBB) scoring and footprint analysis. The effect of MP treatment on BBB score was apparent the day after SCI whereas the effect of NgR(310)ecto-Fc was not apparent until 2 weeks after SCI. NgR(310)ecto-Fc or MP treatment resulted in increased axonal sprouting and/or regeneration, quantified by counting biotin dextran amine-labeled corticospinal tract axons, and increased the number of axons contacting motor neurons in the ventral horn gray matter caudal to the lesion. Combined treatment with NgR(310)ecto-Fc and MP had a more pronounced effect on recovery of function and axonal growth compared with either treatment alone. The data demonstrate that NgR(310)ecto-Fc and MP act in a temporally and mechanistically distinct manner and suggest that they may have complementary effects.


Subject(s)
Methylprednisolone/therapeutic use , Receptors, Peptide/therapeutic use , Spinal Cord Injuries/drug therapy , Analysis of Variance , Animals , Axons/drug effects , Axons/physiology , Behavior, Animal , Biotin/analogs & derivatives , Biotin/metabolism , Cells, Cultured , Chick Embryo , Dextrans/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Drug Therapy, Combination , Exploratory Behavior/drug effects , Female , GPI-Linked Proteins , Ganglia, Spinal/cytology , Immunoglobulin G/therapeutic use , Laminectomy/methods , Myelin Proteins , Myelin Sheath/metabolism , Nerve Regeneration/drug effects , Neurons/drug effects , Neurons/physiology , Nogo Receptor 1 , Pyramidal Tracts/drug effects , Pyramidal Tracts/metabolism , Rats , Rats, Long-Evans , Receptors, Cell Surface , Receptors, Peptide/biosynthesis , Receptors, Peptide/chemistry , Receptors, Peptide/immunology , Recombinant Proteins/therapeutic use , Recovery of Function/drug effects , Spinal Cord Injuries/physiopathology
13.
J Cereb Blood Flow Metab ; 25(8): 978-89, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15815587

ABSTRACT

Pharmacological studies using bradykinin B2 receptor antagonists suggest that bradykinin, an early mediator of inflammation and the main metabolite of the kallikrein-kinin system, is involved in secondary brain damage after cerebral ischemia. However, the time-course of bradykinin production and kinin receptor expression as well as the conclusive role of bradykinin B2 receptors for brain damage after experimental stroke have not been elucidated so far. C57/Bl6 mice were subjected to 45 mins of middle cerebral artery occlusion (MCAO) and 2, 4, 8, 24, and 48 h later brains were removed for the analysis of tissue bradykinin concentration and kinin B2 receptor mRNA and protein expression. Brain edema, infarct volume, functional outcome, and long-term survival were assessed in WT and B2-/- mice 24 h or 7 days after MCAO. Tissue bradykinin was maximally increased 12 h after ischemia (three-fold), while kinin B2 receptor mRNA upregulation peaked 24 to 48 h after MCAO (10- to 12-fold versus naïve brain tissue). Immunohistochemistry revealed that kinin B2 receptors were constitutively and widely expressed in mouse brain, were upregulated 2 h after ischemia in cells showing signs of ischemic damage, and remained upregulated in the penumbra up to 24 h after ischemia. B2-/- mice had improved motor function (P<0.05), smaller infarct volumes (-38%; P<0.01), developed less brain edema (-87%; P<0.05), and survived longer (P<0.01) as compared with wild-type controls. The current results show that bradykinin is produced in the brain, kinin B2 receptors are upregulated on dying cells, and B2 receptors are involved in cell death and brain edema formation after experimental stroke.


Subject(s)
Bradykinin/metabolism , Brain Chemistry/physiology , Brain Edema/pathology , Brain Ischemia/pathology , Receptor, Bradykinin B2/biosynthesis , Animals , Blotting, Western , Bradykinin/blood , Cell Death/physiology , Cerebral Infarction/pathology , Immunohistochemistry , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , RNA, Messenger/biosynthesis , Receptor, Bradykinin B2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
14.
J Neurosci ; 24(46): 10511-20, 2004 Nov 17.
Article in English | MEDLINE | ID: mdl-15548666

ABSTRACT

The growth of injured axons in the adult mammalian CNS is limited after injury. Three myelin proteins, Nogo, MAG (myelin-associated glycoprotein), and OMgp (oligodendrocyte myelin glycoprotein), bind to the Nogo-66 receptor (NgR) and inhibit axonal growth in vitro. Transgenic or viral blockade of NgR function allows axonal sprouting in vivo. Here, we administered the soluble function-blocking NgR ectodomain [aa 27-310; NgR(310)ecto] to spinal-injured rats. Purified NgR(310)ecto-Fc protein was delivered intrathecally after midthoracic dorsal over-hemisection. Axonal sprouting of corticospinal and raphespinal fibers in NgR(310)ecto-Fc-treated animals correlates with improved spinal cord electrical conduction and improved locomotion. The ability of soluble NgR(310)ecto to promote axon growth and locomotor recovery demonstrates a therapeutic potential for NgR antagonism in traumatic spinal cord injury.


Subject(s)
Axons/physiology , Myelin Proteins/antagonists & inhibitors , Myelin-Associated Glycoprotein/antagonists & inhibitors , Myelin-Associated Glycoprotein/metabolism , Receptors, Peptide/physiology , Spinal Cord Injuries/pathology , Animals , Axons/metabolism , Evoked Potentials, Motor , Female , GPI-Linked Proteins , Injections, Spinal , Motor Activity , Myelin-Oligodendrocyte Glycoprotein , Nogo Proteins , Nogo Receptor 1 , Oligodendroglia/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Serotonin/metabolism , Solubility , Spinal Cord/physiopathology , Spinal Cord/ultrastructure , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
15.
Nat Neurosci ; 7(3): 221-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14966521

ABSTRACT

Axon regeneration in the adult CNS is prevented by inhibitors in myelin. These inhibitors seem to modulate RhoA activity by binding to a receptor complex comprising a ligand-binding subunit (the Nogo-66 receptor NgR1) and a signal transducing subunit (the neurotrophin receptor p75). However, in reconstituted non-neuronal systems, NgR1 and p75 together are unable to activate RhoA, suggesting that additional components of the receptor may exist. Here we describe LINGO-1, a nervous system-specific transmembrane protein that binds NgR1 and p75 and that is an additional functional component of the NgR1/p75 signaling complex. In non-neuronal cells, coexpression of human NgR1, p75 and LINGO-1 conferred responsiveness to oligodendrocyte myelin glycoprotein, as measured by RhoA activation. A dominant-negative human LINGO-1 construct attenuated myelin inhibition in transfected primary neuronal cultures. This effect on neurons was mimicked using an exogenously added human LINGO-1-Fc fusion protein. Together these observations suggest that LINGO-1 has an important role in CNS biology.


Subject(s)
Membrane Proteins/metabolism , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Regeneration/physiology , Receptors, Cell Surface/metabolism , Receptors, Nerve Growth Factor/metabolism , Amino Acid Sequence/genetics , Animals , Animals, Newborn , Astrocytes/metabolism , Axons/metabolism , Base Sequence/genetics , Cells, Cultured , DNA, Complementary/analysis , DNA, Complementary/genetics , Fetus , GPI-Linked Proteins , Humans , Macromolecular Substances , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Molecular Sequence Data , Mutation/genetics , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/isolation & purification , Myelin-Oligodendrocyte Glycoprotein , Nerve Tissue Proteins , Nogo Receptor 1 , Protein Structure, Tertiary/genetics , Rats , Receptor, Nerve Growth Factor , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...