Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
2.
Methods Mol Biol ; 2225: 227-239, 2021.
Article in English | MEDLINE | ID: mdl-33108666

ABSTRACT

Severe inflammatory disease initiated by neurotrauma and stroke is of primary concern in these intractable pathologies as noted in recent studies and understanding of the pathogenesis of spinal cord injury (SCI) in the rat model. Successful anti-inflammatory treatments should result in neuroprotection and limit the loss of neurological function to injury caused by the initial damage. Continuous subdural infusion offers direct access to the cavity of injury (COI) that forms after balloon crush SCI deep in the spinal cord. Some anti-inflammatory compounds are not likely capable of crossing the blood-spinal cord barrier. Subdural infusion of myxoma virus-derived Serp-1, an anti-thrombotic/anti-thrombolytic, and also of M-T7, a chemokine inhibitor, improved the locomotor scores and pain sensation scores as well as reduced the numbers of macrophages in the COI by 50 and 80%, respectively, while intraperitoneal infusion of either protein had little effect. Injection of a chitosan hydrogel loaded with Serp-1 into the dorsal spinal column crush also resulted in improved neurological deficits and in reduction of the size of the crush lesion 4 weeks after injury. While neurological scores in a simplified hind-end (HE) locomotor test together with a toe-pinch withdrawal test demonstrated improvement in all balloon crush injury and dorsal spinal crush injury rats, a severe inflammation is induced by the injury indicating additional damage to the spinal cord. Thus neurological function testing can be contradictory, rather than corresponding, to the pathogenesis of SCI. The count of macrophages in the COI offers a precise, reliable method of measuring the effectiveness of a neuroprotective treatment of SCI in preclinical studies.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Immunologic Factors/pharmacology , Myxoma virus/chemistry , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/drug therapy , Viral Proteins/pharmacology , Animals , Anti-Inflammatory Agents/immunology , Chitosan/chemistry , Disease Models, Animal , Drug Delivery Systems/methods , Hydrogels/chemistry , Immunologic Factors/immunology , Injections, Epidural , Macrophages/drug effects , Macrophages/immunology , Male , Motor Activity/drug effects , Neuroprotective Agents/immunology , Rats , Rats, Long-Evans , Receptors, Interferon/immunology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology , Viral Proteins/immunology
3.
Mediators Inflamm ; 2020: 8191079, 2020.
Article in English | MEDLINE | ID: mdl-33061831

ABSTRACT

Disrupted immune response is an important feature of many neurodegenerative conditions, including sepsis-associated cognitive impairment. Accumulating evidence has demonstrated that immune memory occurs in microglia, which has a significant impact on pathological hallmarks of neurological diseases. However, it remains unclear whether immune memory can cause subsequent alterations in the brain immune response and affect neurobehavioral outcomes in sepsis survivors. In the present study, mice received daily intraperitoneal injection of low-dose lipopolysaccharide (LPS, 0.1 mg/kg) for three consecutive days to induce immune memory (immune tolerance) and then were subjected to sham operation or cecal ligation and puncture (CLP) 9 months later, followed by a battery of neurobehavioral and biochemical studies. Here, we showed that repeated low-dose LPS injection-induced immune memory protected mice from sepsis-induced cognitive and affective impairments, which were accompanied by significantly decreased brain proinflammatory cytokines and immune response. In conclusion, our study suggests that modulation of brain immune responses by repeated LPS injections confers neuroprotective effects by preventing overactivated immune response in response to subsequent septic insult.


Subject(s)
Immunity, Innate/physiology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Neuroprotective Agents/immunology , Sepsis/immunology , Animals , Cecum/injuries , Cytokines/blood , Fluorescent Antibody Technique , Ligation/adverse effects , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Open Field Test , Punctures/adverse effects , Sepsis/blood
4.
Int Immunopharmacol ; 77: 105970, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31675618

ABSTRACT

Neutrophils have been traditionally considered as the major mediators of harmful inflammatory responses in ischemic stroke, whereas accumulating evidence indicates that neutrophils can be polarized into an N2 phenotype. Similar to M2 microglia, N2 neutrophils contribute to resolution of inflammation and may participate in neuroprotection. However, it remains unclear whether N2 neutrophils protect ischemic neurons and whether they are associated with long-term outcomes after transient cerebral ischemia in rats. The present study proved that N2 neutrophils protected against oxygen glucosedeprivation/re-oxygenation (OGD/R)-induced primary cortical neuron injury via brain-derived neurotrophic factor/tropomyosin-related kinase B (BDNF/TrkB) signaling. In addition, in vivo studies revealed that transient middle cerebral artery occlusion (tMCAO)-induced injury exhibited spontaneous recovery over time in rats. Moreover, neutrophils could infiltrate the ipsilateral brain parenchyma from the periphery after transient cerebral ischemia. Pearson's correlation analysis indicated that the proportion of N2 neutrophils in ipsilateral brain parenchyma was negatively correlated with the number of degenerating neurons, modified Neurological Severity Score (mNSS), brain water content and infarct volume, and positively correlated with the number of surviving neurons and grip strength. In summary, the present study shows that N2 neutrophils likely participate in spontaneous recovery after transient cerebral ischemia by inhibiting ischemic neuron damage in rats, which indicates that N2 neutrophils may represent promising therapeutic target for promoting recovery after ischemic stroke.


Subject(s)
Brain Ischemia/immunology , Ischemic Attack, Transient/immunology , Neurons/immunology , Neutrophils/immunology , Animals , Brain/immunology , Cell Survival/immunology , Disease Models, Animal , Infarction, Middle Cerebral Artery/immunology , Male , Membrane Glycoproteins/immunology , Microglia/immunology , Neuroprotection/immunology , Neuroprotective Agents/immunology , Rats , Rats, Sprague-Dawley , Receptor, trkB/immunology , Signal Transduction/immunology , Stroke/immunology
6.
Genet Mol Res ; 16(1)2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28340277

ABSTRACT

This study aimed to evaluate the immunomodulatory and neuroprotective effects of allogeneic and cryopreserved mesenchymal stem cells (MSCs) on spinal cord injury. A total of 120 rats were distributed into the following groups: negative control (NC) - without injury, positive control (PC) - with injury without treatment, and group treated with MSC (GMSC) - with injury and treated. Motor function was evaluated by the BBB test at 24, 48, and 72 h and at 8 and 21 postoperative days. Spinal cords were evaluated by histopathology and immunohistochemistry to determine the expression of CD68, NeuN, and GFAP. IL-10, TNF-α, IL-1ß, TGF-ß, BDNF, GDNF, and VEGF expression was quantified by RT-PCR. The GMSC presented higher scores for motor function at 72 h and 8 and 21 days after injury, lower expression of CD68 at 8 days, and lower expression of GFAP at 21 days compared to the PC. In addition, higher expression of NeuN and lower degeneration of the white matter occurred at 21 days. The GMSC also showed higher expression of IL-10 24 h after injury, GDNF at 48 h and 8 days, and VEGF at 21 days. Moreover, lower expression of TNF-α was observed at 8 and 21 days and TGF-ß at 24 h and 21 days. There were no differences in the expression of IL-1ß and BDNF between the GMSC and PC. Thus, cryopreserved MSCs promote immunomodulatory and neuroprotective effects in rats with spinal cord injury by increasing IL-10, GDNF, and VEGF expression and reducing TNF-α and TGF-ß expression.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Regenerative Medicine/methods , Spinal Cord Injuries/therapy , Animals , Cryopreservation/methods , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Interleukin-10/metabolism , Male , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Neuroprotective Agents/immunology , Rats , Rats, Inbred Lew , Spinal Cord Injuries/immunology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Mol Med Rep ; 14(2): 1574-86, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27357155

ABSTRACT

The Bacillus Calmette-Guérin (BCG) vaccine is routinely administered to human neonates worldwide. BCG has recently been identified as a neuroprotective immune mediator in several neuropathological conditions, exerting neuroprotection in a mouse model of Parkinson's disease and slowing the progression of clinically isolated syndrome in patients with multiple sclerosis. The immune system is significantly involved in brain development, and several types of neonatal immune activations exert influences on the brain and behavior following a secondary immune challenge in adulthood. However, whether the neonatal BCG vaccination affects the brain in adulthood remains to be elucidated. In the present study, newborn C57BL/6 mice were injected subcutaneously with BCG (105 colony forming units) or phosphate­buffered saline (PBS). A total of 12 weeks later, the mice were injected intraperitoneally with 330 µg/kg lipopolysaccharide (LPS) or PBS. The present study reported that the neonatal BCG vaccination alleviated sickness, anxiety and depression­like behavior, lessened the impairments in hippocampal cell proliferation and downregulated the proinflammatory responses in the serum and brain that were induced by the adult LPS challenge. However, BCG vaccination alone had no evident influence on the brain and behavior in adulthood. In conclusion, the neonatal BCG vaccination alleviated the neurobehavioral impairments and neuroinflammation induced by LPS exposure in adult mice, suggesting a potential neuroprotective role of the neonatal BCG vaccination in adulthood.


Subject(s)
BCG Vaccine/immunology , Behavior, Animal , Immunization , Inflammation/physiopathology , Nervous System Diseases/physiopathology , Neuroprotective Agents/immunology , Age Factors , Animals , Animals, Newborn , Anxiety , Brain/metabolism , Brain/physiopathology , Cytokines/metabolism , Depression , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/physiopathology , Inflammation/etiology , Inflammation/therapy , Inflammation Mediators/metabolism , Lipopolysaccharides/adverse effects , Mice , Nervous System Diseases/etiology , Nervous System Diseases/therapy , Polysaccharides/metabolism , Serotonin/metabolism
8.
Science ; 353(6294): aaf3646, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27365452

ABSTRACT

Regenerative therapies are limited by unfavorable environments in aging and diseased tissues. A promising strategy to improve success is to balance inflammatory and anti-inflammatory signals and enhance endogenous tissue repair mechanisms. Here, we identified a conserved immune modulatory mechanism that governs the interaction between damaged retinal cells and immune cells to promote tissue repair. In damaged retina of flies and mice, platelet-derived growth factor (PDGF)-like signaling induced mesencephalic astrocyte-derived neurotrophic factor (MANF) in innate immune cells. MANF promoted alternative activation of innate immune cells, enhanced neuroprotection and tissue repair, and improved the success of photoreceptor replacement therapies. Thus, immune modulation is required during tissue repair and regeneration. This approach may improve the efficacy of stem-cell-based regenerative therapies.


Subject(s)
Immunomodulation , Nerve Growth Factors/immunology , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Retina/physiology , Wound Healing/immunology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Egg Proteins/metabolism , Evolution, Molecular , Gene Expression Profiling , Hemocytes/immunology , Immunity, Innate , Mice , Mice, Inbred C57BL , Models, Animal , Nerve Growth Factors/genetics , Neuroprotective Agents/immunology , Photoreceptor Cells, Invertebrate/drug effects , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Platelet-Derived Growth Factor/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Retina/drug effects , Retina/injuries , Retinal Degeneration/therapy , Signal Transduction , Wound Healing/drug effects
9.
Front Biosci (Landmark Ed) ; 21(5): 958-72, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27100484

ABSTRACT

Interphotoreceptor retinoid-binding protein (IRBP) is a classic inducer of experimental autoimmune uveoretinitis (EAU). Although IRBP causes neuronal loss in susceptible animals, resistant animals such as Sprague-Dawley (SPD) rats can benefit from the evoked protective autoimmune responses. The aim of the present study was to analyze the neuroprotective effects of IRBP against light-induced photoreceptor degeneration. We immunized 75 male SPD rats with IRBP and the rats were then exposed to blue light for 24 hours (IRBP group). Seventy five rats were included in the control group. We found that the number of apoptotic cells in the outer nuclear layer (ONL) peaked on 1 day after light exposure, and the ONL thickness decreased significantly on day 3. OX42-positive cells appeared in the ONL immediately after light exposure, and their number peaked on day 3, and changed from resting ramified cells to activated amoeboid cells. Compared with the control group (n=75), the IRBP group showed less apoptotic cells, a thicker ONL, and reduced expression of tumor necrosis factor-alpha. These outcomes indicate the IRPB might protect retinal photoreceptors against light-induced injury.


Subject(s)
Light/adverse effects , Peptide Fragments/immunology , Photoreceptor Cells, Vertebrate/immunology , Photoreceptor Cells, Vertebrate/radiation effects , Retinol-Binding Proteins/immunology , Animals , Apoptosis/immunology , Apoptosis/radiation effects , Autoimmune Diseases/etiology , Autoimmune Diseases/prevention & control , Disease Models, Animal , Male , Microglia/immunology , Microglia/pathology , Microglia/radiation effects , Neuroprotective Agents/immunology , Photoreceptor Cells, Vertebrate/pathology , Rats , Rats, Sprague-Dawley , Retinitis/etiology , Retinitis/prevention & control , Tumor Necrosis Factor-alpha/metabolism , Uveitis/etiology , Uveitis/prevention & control , Vaccination
10.
Brain Behav ; 6(4): e00449, 2016 04.
Article in English | MEDLINE | ID: mdl-27127723

ABSTRACT

BACKGROUND: Stroke could lead to serious morbidity, of which ischemic stroke counts for majority of the cases. Inflammation plays an important role in the pathogenesis of ischemic stroke, thus drugs targeting inflammation could be potentially neuroprotective. Estradiol was shown to be neuroprotective as well as anti-inflammatory in animal models of ischemic stroke with unclear mechanism. We hypothesize that the anti-inflammatory and neuroprotective effect of estradiol is mediated by the estradiol receptor G protein-coupled estrogen receptor 1 (GPER) expressed on microglia. METHODS: We have generated the rat global cerebral ischemic model and the primary microglia culture to study the neuroprotective and anti-inflammatory effect of estradiol. We have further used pharmacological methods and siRNA knockdown approach to study the underlying mechanism. RESULTS: We found that estradiol reduced the level of proinflammatory cytokines including IL-1ß and TNF-α, both in vivo and in vitro. We also found that the specific GPER agonist G1 could reduce the level of IL-1ß (P = 0 P = 0.0017, one-way ANOVA and post hoc test) and TNF-α (P < 0.0001) in the primary microglia culture. Moreover, the specific GPER antagonist G15 was able to abolish the anti-inflammatory effect of estradiol. Estradiol failed to reduce the level of IL-1ß (P = 0.4973, unpaired Student's t-test) and TNF-α (P = 0.1627) when GPER was knocked down. CONCLUSIONS: Our studies have suggested that GPER expressed on microglia mediated the anti-inflammatory effect of estradiol after ischemic stroke. Our studies could potentially help to develop more specific drugs to manage inflammation postischemic stroke.


Subject(s)
Estradiol/metabolism , Inflammation , Microglia , Neuroprotective Agents , Receptors, G-Protein-Coupled , Stroke , Animals , Brain Ischemia/immunology , Brain Ischemia/metabolism , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/metabolism , Microglia/immunology , Microglia/metabolism , Neuroprotective Agents/immunology , Neuroprotective Agents/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Stroke/immunology , Stroke/metabolism
11.
Hum Mol Genet ; 25(2): 317-27, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26604138

ABSTRACT

Neuroinflammation, immune reactivity and mitochondrial abnormalities are considered as causes and/or contributors to neuronal degeneration. Peroxisome proliferator-activated receptors (PPARs) regulate both inflammatory and multiple other pathways that are implicated in neurodegeneration. In the present study, we investigated the efficacy of fenofibrate (Tricor), a pan-PPAR agonist that activates PPAR-α as well as other PPARs. We administered fenofibrate to superoxide dismutase 1 (SOD1(G93A)) mice daily prior to any detectable phenotypes and then animal behavior, pathology and longevity were assessed. Treated animals showed a significant slowing of the progression of disease with weight loss attenuation, enhanced motor performance, delayed onset and survival extension. Histopathological analysis of the spinal cords showed that neuronal loss was significantly attenuated in fenofibrate-treated mice. Mitochondria were preserved as indicated by Cytochrome c immunostaining in the spinal cord, which maybe partly due to increased expression of the PPAR-γ co-activator 1-α. The total mRNA analysis revealed that neuroprotective and anti-inflammatory genes were elevated, while neuroinflammatory genes were down-regulated. This study demonstrates that the activation of PPAR-α action via fenofibrate leads to neuroprotection by both reducing neuroinflammation and protecting mitochondria, which leads to a significant increase in survival in SOD1(G93A) mice. Therefore, the development of therapeutic strategies to activate PPAR-α as well as other PPARs may lead to new therapeutic agents to slow or halt the progression of amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Fenofibrate/pharmacology , Inflammation/metabolism , Neurons/physiology , PPAR alpha/agonists , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Death , Disease Progression , Female , Fenofibrate/immunology , Inflammation/drug therapy , Male , Mice , Mice, Transgenic , Mitochondria/drug effects , Neurons/drug effects , Neurons/immunology , Neuroprotective Agents/immunology , Neuroprotective Agents/pharmacology , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/physiopathology
12.
Curr Pharm Biotechnol ; 16(6): 492-505, 2015.
Article in English | MEDLINE | ID: mdl-25860061

ABSTRACT

Regeneration in the central nervous system (CNS) of adult mammalian after traumatic injury is limited, which often causes permanent functional motor and sensory loss. After spinal cord injury (SCI), the lack of regeneration is mainly attributed to the presence of a hostile microenvironment, glial scarring, and cavitation. Besides, inflammation has also been proved to play a crucial role in secondary degeneration following SCI. The more prominent treatment strategies in experimental models focus mainly on drugs and cell therapies, however, only a few strategies applied in clinical studies and therapies still have only limited effects on the repair of SCI. Recently, the interests in immunotherapy strategies for CNS are increasing in number and breadth. Immunotherapy strategies have made good progresses in treating many CNS degenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), stroke, and multiple sclerosis (MS). However, the strategies begin to be considered to the treatment of SCI and other neurological disorders in recent years. Besides anti-inflamatory therapy, immunization with protein vaccines and DNA vaccines has emerged as a novel therapy strategy because of the simplicity of preparation and application. An inflammatory response followed by spinal cord injury, and is controled by specific signaling molecules, such as some cytokines playing a crucial role. As a result, appropriate immunoregulation, the expression of pro-inflammatory cytokines and anti-inflammatory cytokines may be an effective therapy strategy for earlier injury of spinal cord. In addition, myelinassociated inhibitors (MAIs) in the injured spinal cord, such as Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte- myelin glycoprotein (OMgp) are known to prevent axonal regeneration through their co-receptors, and to trigger demyelinating autoimmunity through T cell-mediated harmful autoimmune response. The antagonism of the MAIs through vaccinating with protein or DNA vaccines targeting Nogo, MAG, OMgp, and their co-receptors, may be an effective strategy for the treatment of SCI. However, immunotherapy such as anti-inflammtory therapy or vaccine targeting MAIs or their receptors, accompanied with the potential in risking autoimmune diseases. As a result, in order to optimize the anti-inflammtory therapy and design of protein or DNA vaccines for their use in the future clinical application, we need to further understand the possible mechanisms of neuroprotective immunity. This review presents recent advances in the development of immunotherapy strategies for the treatment of axonal degeneration and demyelination, and improvement of motor function after SCI.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunization/methods , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/immunology , Spinal Cord Injuries/therapy , Spinal Cord/immunology , Antibodies, Monoclonal/immunology , Humans , Neuroprotective Agents/immunology , Spinal Cord/drug effects
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(4): 437-42, 2015 Apr.
Article in Chinese | MEDLINE | ID: mdl-25854558

ABSTRACT

OBJECTIVE: To explore the neuroprotective effects of preconditioning with low-dose lipopolysaccharide (LPS) in rats after spinal cord injury and its possible mechanism. METHODS: Forty-eight female SD rats were randomly divided into four groups as follows: empty virus (EV), LPS combined with empty virus (LPS-EV), nuclear factor erythroid 2-related factor 2 (Nrf2) interference virus (NIV), and LPS combined with Nrf2 interference virus (LPS-NIV) (n=12 per group). The model of traumatic spinal cord injury (TSCI) was established by the modified Allen's method. Hind limb motor function of the rats was assessed by the Basso, Beattie and Bresnahan (BBB) score 3 days after the operation. The injured spinal cord tissues were harvested after the operation. The pathological changes of spinal cord were observed by HE staining. The Nissl body and neuron survival index were assessed by Nissl staining. The expressions of Nrf2, NF-κB and associated pro-inflammatory cytokines (IL-1ß, TNF-α) were detected by immunohistochemical staining and Western blotting. RESULTS: NIV group showed descended Nrf2 protein expression and ascended NF-κB, IL-1ß, TNF-α protein levels compared with EV group, and no significant difference from LPS-NIV group. The Nrf2 protein expression of LPS-EV group increased significantly compared with EV group and LPS-NIV group, and NF-κB, TNF-α and IL-1ß protein expression decreased significantly at the same time. Compared with those of EV group and LPS-NIV group, the neuron survival index of LPS-EV group was improved. The morphological change of LPS-EV group was also obviously alleviated. The BBB score had no statistical significance among these groups. CONCLUSION: Low-dose LPS preconditioning had neuroprotective effects on spinal cord injury. This protective effect was mediated by activating the Nrf2 to down-regulate expressions of NF-κB and pro-inflammatory cytokines and alleviate inflammatory response.


Subject(s)
Lipopolysaccharides/immunology , NF-E2-Related Factor 2/immunology , Spinal Cord Injuries/genetics , Animals , Female , Genetic Therapy , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , NF-kappa B/immunology , Neuroprotective Agents/immunology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/immunology , Spinal Cord Injuries/therapy , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Up-Regulation
14.
Mol Immunol ; 60(2): 109-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24815539

ABSTRACT

Intracerebral hemorrhage (ICH) causes morbidity and mortality and commonly follows the reperfusion after an ischemic event. Microglial activation mediated cytokine and protease secretion contributes to brain injury in ICH. Previous studies have shown that sinomenine possesses potent immunoregulatory properties. However, little is known about its exact role in ICH. In the present study, to investigate the effect of sinomenine on microglial cells inflammation, we treated ICH-challenged BV2 microglial cells with sinomenine in vitro, and explored its neuroprotection role in intracerebral hemorrhage in vivo. Changes in inflammatory cytokines, such as TNF-α, IL-1ß and IL-6, reactive oxygen species (ROS) and NF-κB activation NF-κB were observed. In addition, the neurological deficit and cerebral water content of ICH mice were studied. The results demonstrated that sinomenine could inhibit the release of these cytokines and attenuate ROS production in a dose-dependent manner, and reduce NF-κB activation. Furthermore, sinomenine markedly inhibited cerebral water content and neurological deficit. In conclusion, our findings suggest that sinomenine played the protective effects through inhibition of microglial inflammation, and the findings also provided a novel therapy to treat ICH induced brain injury.


Subject(s)
Brain Injuries/drug therapy , Cerebral Hemorrhage/drug therapy , Microglia/drug effects , Morphinans/pharmacology , Animals , Brain Injuries/immunology , Cell Line , Cell Movement/drug effects , Cell Movement/immunology , Cerebral Hemorrhage/immunology , Inflammation/drug therapy , Inflammation/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , NF-kappa B/immunology , Neuroprotective Agents/immunology , Neuroprotective Agents/pharmacology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology
15.
Pharmacol Res ; 86: 32-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24844438

ABSTRACT

The existence of acylethanolamides (AEs) in the mammalian brain has been known for decades. Among AEs, palmitoylethanolamide (PEA) is abundant in the central nervous system (CNS) and conspicuously produced by neurons and glial cells. Antihyperalgesic and neuroprotective properties of PEA have been mainly related to the reduction of neuronal firing and to control of inflammation. Growing evidence suggest that PEA may be neuroprotective during CNS neurodegenerative diseases. Advances in the understanding of the physiology and pharmacology of PEA have potentiated its interest as useful biological tool for disease management. Several rapid non-genomic and delayed genomic mechanisms of action have been identified for PEA as peroxisome proliferator-activated receptor (PPAR)-α dependent. First, an early molecular control, through Ca(+2)-activated intermediate- and/or big-conductance K(+) channels opening, drives to rapid neuronal hyperpolarization. This is reinforced by the increase of the inward Cl(-) currents due to the modulation of the gamma aminobutyric acid A receptor and by the desensitization of the transient receptor potential channel type V1. Moreover, the gene transcription-mediated mechanism sustains the long-term anti-inflammatory effects, by reducing pro-inflammatory enzyme expression and increasing neurosteroid synthesis. Overall, the integration of these different modes of action allows PEA to exert an immediate and prolonged efficacious control in neuron signaling either on inflammatory process or neuronal excitability, maintaining cellular homeostasis. In this review, we will discuss the effect of PEA on metabolism, behavior, inflammation and pain perception, related to the control of central functions and the emerging evidence demonstrating its therapeutic efficacy in several neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ethanolamines/pharmacology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Palmitic Acids/pharmacology , Amides , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/therapeutic use , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/metabolism , Ethanolamines/immunology , Ethanolamines/metabolism , Ethanolamines/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/immunology , Neuroprotective Agents/metabolism , Neuroprotective Agents/therapeutic use , PPAR alpha/immunology , PPAR alpha/metabolism , Pain/drug therapy , Pain/immunology , Pain/metabolism , Palmitic Acids/immunology , Palmitic Acids/metabolism , Palmitic Acids/therapeutic use
16.
Neurobiol Learn Mem ; 107: 50-64, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24239620

ABSTRACT

Epidemiological studies demonstrated association between head injury (HI) and the subsequent development of Alzheimer's disease (AD). Certain hallmarks of AD, e.g. amyloid-ß (Aß) containing deposits, may be found in patients following traumatic BI (TBI). Recent studies uncover the cellular prion protein, PrP(C), as a receptor for soluble polymeric forms of Aß (sAß) which are an intermediate of such deposits. We aimed to test the hypothesis that targeting of PrP(C) can prevent Aß related spatial memory deficits in olfactory bulbectomized (OBX) mice utilized here to resemble some clinical features of AD, such as increased level of Aß, memory loss and deficit of the CNS cholin- and serotonin-ergic systems. We demonstrated that immunization with the a.a. 95-123 fragment of cellular prion (PrP-I) recovered cortical and hippocampus neurons from OBX induced degeneration, rescued spatial memory loss in Morris water maze test and significantly decrease the Aß level in brain tissue of these animals. Affinity purified anti-PrP-I antibodies rescued pre-synaptic biomarker synaptophysin eliciting similar effect on memory of OBX mice, and protected hippocampal neurones from Aß25-35-induced toxicity in vitro. Immunization OBX mice with a.a. 200-213 fragment of cellular prion (PrP-II) did not reach a significance in memory protection albeit having similar to PrP-I immunization impact on Aß level in brain tissue. The observed positive effect of targeting the PrP-I by either active or passive immunization on memory of OBX mice revealed the involvement of the PrP(C) in AD-like pathology induced by olfactory bulbectomy. This OBX model may be a useful tool for mechanistic and preclinical therapeutic investigations into the association between PrP(C) and AD.


Subject(s)
Memory Disorders/therapy , Nerve Degeneration/therapy , Neuroprotective Agents/immunology , Peptide Fragments/immunology , PrPC Proteins/immunology , Prions/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibodies/immunology , Hippocampus/pathology , Immunization , Immunization, Passive , Male , Maze Learning/physiology , Mice , Mice, Inbred Strains , Neurons/pathology , Olfactory Bulb/pathology , Rats , Rats, Sprague-Dawley , Temporal Lobe/pathology
17.
Neuropharmacology ; 76 Pt A: 184-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23831681

ABSTRACT

It is well established that immunosurveillance is active in the CNS and plays a key role in several CNS disorders but the exact role of immune cells remains elusive. Thus, in the present study we investigated whether lymphocytes are protective/detrimental in in vitro models of excitotoxicty. Kainate (KA)-induced neuronal death was significantly reduced following exposure to mixed lymphocytes or purified T lymphocytes containing either activated or non-activated T-lymphocytes. Conditioned media from lymphocyte preparations, but not boiled conditioned media, was protective against KA-induced toxicity indicating soluble mediators underlie the observed neuroprotection with cytokine arrays indicating IL-16 as the likely candidate. A role for astrocytes was established as the neuroprotection was abolished in the presence of the glial toxin, fluoroacetate. Furthermore, lymphocytes inhibited p38 MAPK and ERK signalling pathways with pharmacological inhibition of these pathways mimicking the protective effect of lymphocytes. Similarly, lymphocytes were neuroprotective against oxygen-glucose deprivation (OGD)-induced cell death with the inhibition of p38 MAPK and ERK signalling pathways involved. These data indicate that lymphocytes are neuroprotective under our experimental conditions and we suggest that astrocytic activation and inhibition of MAPK signalling cascades are involved but further studies are required to investigate whether similar mechanisms underlie the actions of lymphocytes in in vivo experimental models of disease. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.


Subject(s)
Astrocytes/immunology , Hippocampus/immunology , Kainic Acid/toxicity , MAP Kinase Signaling System/immunology , Nerve Degeneration/chemically induced , Nerve Degeneration/immunology , Neuroprotective Agents/immunology , T-Lymphocytes/immunology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Death/drug effects , Cell Death/immunology , Culture Media, Conditioned/pharmacology , Cytokines/immunology , Fluoroacetates/toxicity , Hippocampus/drug effects , Hippocampus/pathology , Lymphocyte Activation/immunology , MAP Kinase Signaling System/drug effects , Mice , Signal Transduction/drug effects , Signal Transduction/immunology
18.
Immunol Lett ; 158(1-2): 95-100, 2014.
Article in English | MEDLINE | ID: mdl-24333341

ABSTRACT

BACKGROUND: Cerebral ischemia is associated with infectious complications due to immunosuppression and decreased T lymphocyte activity. G-CSF, which has neuroprotective properties, is known to modulate inflammatory processes after induced stroke. The aim of our study was to investigate the impact of G-CSF in experimental stroke and to compare two different modes of treatment, focusing on circulating T lymphocytes. METHODS: Cerebral ischemia was induced in Wistar rats by occlusion of the middle cerebral artery, followed by reperfusion after 1h. G-CSF was applied either as a single dose 30 min after occlusion, or daily for seven days. Silver staining was used to determine infarct size. T lymphocytes in the peripheral blood were measured before and 7 days after induced cerebral ischemia by flow cytometry. In addition, migration of CD3-expressing T lymphocytes into the brain was investigated by immunohistochemistry. RESULTS: Both single dose and daily treatment with G-CSF significantly reduced infarct size. A significant improvement of neurological outcome was only observed after single application of G-CSF. While a decrease in peripheral T lymphocytes was detected seven days after induced stroke, no reduction was observed in the G-CSF-treated groups. Apart from that, G-CSF significantly reduced the number of brain migrated T lymphocytes in both treatment settings as compared to vehicle. CONCLUSION: A single dose of G-CSF exerted neuroprotective effects in ischemic stroke, which were less pronounced after daily G-CSF application. Both treatment strategies inhibited stroke-induced reduction of T lymphocytes in peripheral blood, which may have contributed to the reduction of infarct size.


Subject(s)
Brain Ischemia/therapy , Brain/immunology , Granulocyte Colony-Stimulating Factor/administration & dosage , Immunotherapy/methods , Neuroprotective Agents/administration & dosage , Stroke/therapy , T-Lymphocytes/immunology , Animals , Blood Circulation , Brain/pathology , Brain Ischemia/immunology , Cell Movement , Clinical Protocols , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/immunology , Humans , Immunity, Cellular , Male , Middle Cerebral Artery , Neuroprotective Agents/immunology , Rats , Rats, Wistar , Reperfusion , Stroke/immunology
19.
J Parkinsons Dis ; 3(4): 581-91, 2013.
Article in English | MEDLINE | ID: mdl-24270241

ABSTRACT

Misfolded proteins and subsequent protein aggregation appears to underlie a significant fraction of neurodegenerative diseases including Parkinson's disease. One of the neuropathological hallmarks of Parkinson's disease is the presence of α-syn containing intracellular inclusions known as Lewy bodies and Lewy neurites. Intrabodies are antibody fragments that have been engineered to be expressed intracellularly. They can be directed towards specific target antigens present in various subcellular locations, and have shown promise in cancer, HIV, autoimmune diseases, and Huntington's disease. More recently they have been shown to modulate abnormalities caused by aggregated α-syn in cell culture. This mini-review mainly focuses on summarizing structural and cellular effects of intrabodies shown to have affinity for different forms of α-synuclein (monomeric, oligomeric and fibrillar), as well as those exhibiting affinity for particular residues of α-synuclein (e.g., the NAC region, C terminal region).


Subject(s)
Antibodies/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Humans , Lewy Bodies/drug effects , Neuroprotective Agents/immunology , Parkinson Disease/immunology , Protein Engineering/methods , Protein Folding/drug effects , alpha-Synuclein/drug effects
20.
Kathmandu Univ Med J (KUMJ) ; 11(42): 132-8, 2013.
Article in English | MEDLINE | ID: mdl-24096220

ABSTRACT

BACKGROUND: Neurodegenerative disease is a progressive loss of neurons from the central nervous system (CNS). Various conditions have been implicated for such conditions including ageing, inflammation, stress and genetic predisposition. Recently, studies have linked neurodegeneration with inflammation. Some studies have suggested the harmful effect of immune response while others have argued its neuroprotective role in neurodegeneration of the CNS. However, the precise role of inflammation and immune cells in such condition is still not clear. OBJECTIVE: To investigate the role of lymphocytes in neurodegeneration of the CNS and determine the underlying mechanism. METHOD: We have used 4-7 days old mouse pups (C57Bl6) to prepare organotypic slice cultures which were cultured for 13-15 days prior to experiment. To induced cell death kainic acid was used and considered as an in vitro model for neurodegeneration. Lymphocytes were obtained from peripheral lymph nodes of 5-10 weeks old adult mouse which were used in the current study. Propidium iodide was used as a fluorescent dye to determine cell death in brain slice cultures. RESULT: Lymphocytes do not induce cell death in slice cultures in the absence of any toxic insult whereas, after applying toxic insult to the slice cultures using kainic acid, lymphocytes show neuroprotection against such insult. Similarly, purified nonactivated and purified activated T cells along with T cells depleted lymphocyte preparation also exhibit neuroprotection against kainic acid-induced cell death. We further, have demonstrated that the observed neuroprotection is contact-independent and soluble mediators released from lymphocytes are responsible for the observed neuroprotection. Moreover, our study has revealed that soluble mediators exhibiting neuroprotection act via astrocytes. CONCLUSION: Lymphocyte preparations are neuroprotective and the observed neuroprotection is contact-independent. Soluble mediators released from lymphocytes are responsible for the observed neuroprotection.


Subject(s)
Cell Death/drug effects , Lymphocytes/immunology , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Neuroprotective Agents/immunology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , In Vitro Techniques , Inflammation/physiopathology , Kainic Acid/pharmacology , Mice , Neurodegenerative Diseases/drug therapy , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...