Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Stroke ; 53(5): 1802-1812, 2022 05.
Article in English | MEDLINE | ID: mdl-35354299

ABSTRACT

Cerebral ischemia and reperfusion initiate cellular events in brain that lead to neurological disability. Investigating these cellular events provides ample targets for developing new treatments. Despite considerable work, no such therapy has translated into successful stroke treatment. Among other issues-such as incomplete mechanistic knowledge and faulty clinical trial design-a key contributor to prior translational failures may be insufficient scientific rigor during preclinical assessment: nonblinded outcome assessment; missing randomization; inappropriate sample sizes; and preclinical assessments in young male animals that ignore relevant biological variables, such as age, sex, and relevant comorbid diseases. Promising results are rarely replicated in multiple laboratories. We sought to address some of these issues with rigorous assessment of candidate treatments across 6 independent research laboratories. The Stroke Preclinical Assessment Network (SPAN) implements state-of-the-art experimental design to test the hypothesis that rigorous preclinical assessment can successfully reduce or eliminate common sources of bias in choosing treatments for evaluation in clinical studies. SPAN is a randomized, placebo-controlled, blinded, multilaboratory trial using a multi-arm multi-stage protocol to select one or more putative stroke treatments with an implied high likelihood of success in human clinical stroke trials. The first stage of SPAN implemented procedural standardization and experimental rigor. All participating research laboratories performed middle cerebral artery occlusion surgery adhering to a common protocol and rapidly enrolled 913 mice in the first of 4 planned stages with excellent protocol adherence, remarkable data completion and low rates of subject loss. SPAN stage 1 successfully implemented treatment masking, randomization, prerandomization inclusion/exclusion criteria, and blinded assessment to exclude bias. Our data suggest that a large, multilaboratory, preclinical assessment effort to reduce known sources of bias is feasible and practical. Subsequent SPAN stages will evaluate candidate treatments for potential success in future stroke clinical trials using aged animals and animals with comorbid conditions.


Subject(s)
Brain Ischemia , Stroke , Aged , Animals , Brain , Brain Ischemia/therapy , Feasibility Studies , Humans , Infarction, Middle Cerebral Artery/therapy , Male , Mice , Stroke/therapy
2.
Cond Med ; 4(6): 280-284, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35634455

ABSTRACT

Stroke is a leading cause of disability and death worldwide. There is evidence that there is a circadian rhythm in stroke with peak occurrence in the morning (6 to 10 am). However, it is not clear if the size of infarcts and the outcome of stroke also varies during the 24-hour period. We hypothesized that the size of cerebral infarct and outcome from stroke would show circadian variation in a mouse suture occlusion model. Seven to eight-month-old C57BL/6J (n =10-12 mice/group) mice were randomly assigned to undergo middle cerebral artery occlusion (MCAO) for 60 minutes at different time points during the 24h day following zeitgeber time at ZT0, ZT6, ZT12, and Z18. Cerebral blood flow was monitored by Laser Speckle Contrast Imaging at baseline after occlusion, and again at 24h post-occlusion. Neurological deficit was observed by using Bederson score at 24h and 48h. The corner test was used to detect unilateral abnormalities in sensory and motor functions in the stroke mice at 48h. To estimate brain infarction, 2,3,5-tryphenyltetrazolium chloride staining was performed 48h after stroke and the infarct area was quantified using NIH-Image J software. We did not find a significant difference in cerebral blood flow at any time point. There was a significant decrease in neurological deficit as assessed using the Bederson Score from 24h (1.82 ± 1.11) to 48h (1.10 ± 0.12) in the ZT18 (midnight) period (p = 0.0025), however there were no differences between groups at 48h. In the corner test, we found right turn preference significantly higher (p = 0.0348) at noon/ZT06 (9.5 ± 1.06) compared to the fully awake (5.5 ± 4.06) (midnight, ZT18) period and ZT0 (6 am, 4.8 ± 0.97, p = 0.0087). Similarly, the infarction volume was significantly higher (p = 0.0220) during the sleep (ZT06, noon) period (35.22 ± 20.77) than when the ischemic mice were fully awake during the midnight/ZT18 period (15.68 ± 7.54). This is the first report demonstrating that mice have larger infarcts and worse short-term outcomes during their sleep period (noon/ZT06) than during their awake period (midnight/ZT18).

3.
Stroke ; 51(3): 967-974, 2020 03.
Article in English | MEDLINE | ID: mdl-32019481

ABSTRACT

Background and Purpose- Microglia are among the first cells to respond to intracerebral hemorrhage (ICH), but the mechanisms that underlie their activity following ICH remain unclear. IL (interleukin)-15 is a proinflammatory cytokine that orchestrates homeostasis and the intensity of the immune response following central nervous system inflammatory events. The goal of this study was to investigate the role of IL-15 in ICH injury. Methods- Using brain slices of patients with ICH, we determined the presence and cellular source of IL-15. A transgenic mouse line with targeted expression of IL-15 in astrocytes was generated to determine the role of astrocytic IL-15 in ICH. The expression of IL-15 was controlled by a glial fibrillary acidic protein promoter (GFAP-IL-15tg). ICH was induced by intraparenchymal injection of collagenase or autologous blood. Results- In patients with ICH and wild-type mice subjected to experimental ICH, we found a significant upregulation of IL-15 in astrocytes. In GFAP-IL-15tg mice, we found that astrocyte-targeted expression of IL-15 exacerbated brain edema and neurological deficits following ICH. This aggravated ICH injury in GFAP-IL-15tg mice is accompanied by increased microglial accumulation in close proximity to astrocytes in perihematomal tissues. Additionally, microglial expression of CD86, IL-1ß, and TNF-α is markedly increased in GFAP-IL-15tg mice following ICH. Furthermore, depletion of microglia using a colony stimulating factor 1 receptor inhibitor diminishes the exacerbation of ICH injury in GFAP-IL-15tg mice. Conclusions- Our findings identify IL-15 as a mediator of the crosstalk between astrocytes and microglia that exacerbates brain injury following ICH.


Subject(s)
Astrocytes/immunology , Brain Injuries/immunology , Cerebral Hemorrhage/immunology , Interleukin-15/immunology , Microglia/immunology , Aged , Aged, 80 and over , Animals , Astrocytes/pathology , Brain Injuries/etiology , Brain Injuries/genetics , Brain Injuries/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/pathology , Female , Humans , Interleukin-15/genetics , Male , Mice , Mice, Transgenic , Microglia/pathology
4.
Neuropharmacology ; 141: 148-157, 2018 10.
Article in English | MEDLINE | ID: mdl-30172846

ABSTRACT

GABAA and GABAC receptors are both GABA-gated chloride channels with distinct pharmacological properties, mainly in their sensitivity to bicuculline and gabazine. In this study, we found that suramin, a purinergic receptor antagonist, is a novel competitive antagonist selective to GABAA over GABAC receptors. Specifically, suramin antagonized the GABA-induced current and the spontaneous opening current of the wild type α1ß2γ2 GABAA receptor with high-level expression in Xenopus oocytes. The antagonism was concentration dependent with an IC50 that varied depending on the concentration of GABA, and with the lowest IC50 of 0.43 µM when antagonizing the spontaneous current. Thus, its potency is slightly higher than bicuculline on the same GABAA receptor. Suramin also antagonized the mouse native brain GABA receptors micro-transplanted into the Xenopus oocytes with its potency depending on the GABA concentration. In addition, in the presence of two fixed concentrations of suramin, the GABA concentration response of the receptor was shifted to the right without reduction of the maximum current. Thus, our results are consistent with that suramin is a competitive antagonist for the α1ß2γ2 GABAA receptor. Interestingly, the rank order of maximum allosteric inhibition (efficacy) of spontaneous current of the GABAA receptor by three competitive antagonists was suramin > bicuculline > gabazine, similar to the rank order of their molecular weight. In contrast, similar to bicuculline, suramin has much lower potency in antagonizing the GABA-induced current of the ρ1 GABAC receptor. In conclusion, we have identified a novel GABAA receptor competitive antagonist, which is selective to the α1ß2γ2 over ρ1 GABA receptors.


Subject(s)
Binding, Competitive/drug effects , GABA Antagonists/pharmacology , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Suramin/pharmacology , Allosteric Regulation , Animals , Bicuculline/pharmacology , Brain Tissue Transplantation , Dose-Response Relationship, Drug , Mice , Oocytes , Patch-Clamp Techniques , Protein Isoforms/antagonists & inhibitors , Pyridazines/pharmacology , Substrate Specificity , Xenopus , gamma-Aminobutyric Acid/pharmacology
5.
Toxicology ; 406-407: 114-122, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30053495

ABSTRACT

Maternal exposure to di-n-butyl phthalate (DBP) induces hypospadias, but the underlying mechanisms remain elusive. Here we hypothesize that aberrant activation of autophagy and epithelial-mesenchymal transition (EMT) are the leading cause of DBP-related hypospadias. Pregnant rats received DBP orally at a dose of 750 mg/kg/day during gestational days 14-18. In DBP-induced hypospadiac male offspring, immunohistochemistry (IHC) staining and Western blot showed increased expression of autophagy and EMT markers in genital tubercle (GT) tissue compared to the control. In addition, lower testosterone levels and androgen receptor (AR) expression in GT tissue were detected. In vitro studies revealed that impaired AR signaling was involved in DBP-induced autophagy and autophagy activation furthermore promoted EMT in urethral epithelial cells. DBP combined with chloroquine, an autophagy inhibitor, reduced the expression of EMT markers compared with DBP treatment alone, while DBP combined with the autophagy inducer rapamycin elevated the expression of EMT markers. The autophagy-lysosomal pathway inhibitor CQ but not proteasome inhibitor MG-132 rescued the decrease of E-cadherin after DBP treatment, which indicated autophagy-induced E-cadherin degradation contributes to DBP-related EMT. Taken together, our findings show that prenatal exposure to DBP induces abnormal autophagy and EMT that may play important roles in hypospadias development.


Subject(s)
Autophagy/drug effects , Dibutyl Phthalate/toxicity , Epithelial-Mesenchymal Transition/drug effects , Plasticizers/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Urothelium/drug effects , Animals , Autophagy/physiology , Cells, Cultured , Epithelial-Mesenchymal Transition/physiology , Female , Hypospadias/chemically induced , Hypospadias/metabolism , Hypospadias/pathology , Male , Maternal Exposure/adverse effects , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley , Urothelium/metabolism , Urothelium/pathology
6.
Stroke ; 49(6): 1471-1478, 2018 06.
Article in English | MEDLINE | ID: mdl-29695462

ABSTRACT

BACKGROUND AND PURPOSE: Autoimmune responses can occur when antigens from the central nervous system are presented to lymphocytes in the periphery or central nervous system in several neurological diseases. However, whether autoimmune responses emerge after brain ischemia and their impact on clinical outcomes remains controversial. We hypothesized that brain ischemia facilitates the genesis of autoimmunity and aggravates ischemic brain injury. METHODS: Using a mouse strain that harbors a transgenic T-cell receptor to a central nervous system antigen, MOG35-55 (myelin oligodendrocyte glycoprotein) epitope (2D2), we determined the anatomic location and involvement of antigen-presenting cells in the development of T-cell reactivity after brain ischemia and how T-cell reactivity impacts stroke outcome. Transient middle cerebral artery occlusion and photothrombotic stroke models were used in this study. We also quantified the presence and status of T cells from brain slices of ischemic patients. RESULTS: By coupling transfer of labeled MOG35-55-specific (2D2) T cells with tetramer tracking, we show an expansion in reactivity of 2D2 T cells to MOG91-108 and MOG103-125 in transient middle cerebral artery occlusion and photothrombotic stroke models. This reactivity and T-cell activation first occur locally in the brain after ischemia. Also, microglia act as antigen-presenting cells that effectively present MOG antigens, and depletion of microglia ablates expansion of 2D2 reactive T cells. Notably, the adoptive transfer of neuroantigen-experienced 2D2 T cells exacerbates Th1/Th17 responses and brain injury. Finally, T-cell activation and MOG-specific T cells are present in the brain of patients with ischemic stroke. CONCLUSIONS: Our findings suggest that brain ischemia activates and diversifies T-cell responses locally, which exacerbates ischemic brain injury.


Subject(s)
Brain Injuries/immunology , Brain Ischemia/immunology , Stroke/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Brain/immunology , Central Nervous System/immunology , Humans , Infarction, Middle Cerebral Artery/immunology , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology
7.
Stroke Vasc Neurol ; 3(1): 34-41, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29600006

ABSTRACT

Infections occur commonly after stroke and are strongly associated with an unfavourable functional outcome of these patients. Approaches for effective management of poststroke infection remain scarce, presenting an urgent need for preventive anti-infection strategies for patients who have suffered a stroke. Emerging evidence indicates that stroke impairs systemic immune responses and increases the susceptibility to infections, suggesting that the modification of impaired immune defence could be beneficial. In this review, we summarised previous attempts to prevent poststroke infections using prophylactic antibiotics and the current understanding of stroke-induced immunosuppression. Further elucidation of the immune mechanisms of stroke will pave the way to tailored design of new treatment to combat poststroke infection via modifying the immune system.


Subject(s)
Bacterial Infections/immunology , Immunocompromised Host , Opportunistic Infections/immunology , Stroke/immunology , Animals , Anti-Bacterial Agents/administration & dosage , Antibiotic Prophylaxis , Bacterial Infections/diagnosis , Bacterial Infections/microbiology , Bacterial Infections/prevention & control , Host Microbial Interactions , Humans , Opportunistic Infections/diagnosis , Opportunistic Infections/microbiology , Opportunistic Infections/prevention & control , Prognosis , Risk Assessment , Risk Factors , Stroke/diagnosis , Stroke/drug therapy
8.
FASEB J ; 32(6): 3336-3345, 2018 06.
Article in English | MEDLINE | ID: mdl-29401614

ABSTRACT

The activation of microglia and the various substances they produce have been linked to the pathologic development of Parkinson's disease (PD), but the precise role of microglia in PD remains to be defined. The survival of microglia depends on colony-stimulating factor 1 receptor (CSF1R) signaling, and CSF1R inhibition results in rapid elimination of microglia in the central nervous system. Using a mouse PD model induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment, we showed that the depletion of microglia via the CSF1R inhibitor PLX3397 exacerbated the impairment of locomotor activities and the loss of dopaminergic neurons. Further, depletion of microglia augmented the production of inflammatory mediators and infiltration of leukocytes in the brain after MPTP exposure. Microglia depletion-induced aggravation of MPTP neurotoxicity was also seen in lymphocyte-deficient mice. In addition, the depletion of microglia did not affect the production of brain-derived neurotrophic factor, but it dramatically augmented the production of inflammatory mediators by astrocytes after MPTP treatment. Our findings suggest microglia play a protective role against MPTP-induced neuroinflammation and dopaminergic neurotoxicity.-Yang, X., Ren, H., Wood, K., Li, M., Qiu, S., Shi, F.-D., Ma, C., Liu, Q. Depletion of microglia augments the dopaminergic neurotoxicity of MPTP.


Subject(s)
Dopaminergic Neurons/metabolism , Inflammation Mediators/metabolism , Leukocytes/metabolism , MPTP Poisoning/metabolism , Microglia/metabolism , Aminopyridines/pharmacology , Animals , Dopaminergic Neurons/pathology , Leukocytes/pathology , MPTP Poisoning/pathology , Male , Mice , Mice, Knockout , Microglia/pathology , Pyrroles/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
9.
Muscle Nerve ; 57(5): 814-820, 2018 05.
Article in English | MEDLINE | ID: mdl-29193204

ABSTRACT

INTRODUCTION: Agrin is essential for the formation and maintenance of neuromuscular junctions (NMJs). NT-1654 is a C-terminal fragment of mouse neural agrin. In this study, we determined the effects of NT-1654 on the severity of experimental autoimmune myasthenia gravis (EAMG). METHODS: EAMG was induced in female Lewis rats by immunization with the Torpedo acetylcholine receptor (tAChR) and complete Freund's adjuvant (CFA). NT-1654 was dissolved in phosphate-buffered saline (PBS) and injected daily subcutaneously into tAChR immunized rats during the first 10 days after immunization, and then every other day for the following 20 days. RESULTS: We showed that NT-1654 attenuated clinical severity, effectively promoted the clustering of AChRs at NMJs, and alleviated the impairment of NMJ transmission and the reduction of muscle-specific kinase (MuSK) in EAMG rats. DISCUSSION: We demonstrated that NT-1654 attenuated clinical severity, effectively promoted the clustering of AChRs at NMJs, and alleviated the impairment of NMJ transmission and the reduction of muscle-specific kinase (MuSK) in EAMG rats. Muscle Nerve 57: 814-820, 2018.


Subject(s)
Agrin/therapeutic use , Immunization/adverse effects , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/pathology , Peptide Fragments/therapeutic use , Action Potentials/physiology , Agrin/biosynthesis , Agrin/chemistry , Animals , Autoantibodies/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Electromyography , Female , Freund's Adjuvant/toxicity , Gene Expression Regulation/drug effects , Membrane Glycoproteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/therapy , Nerve Tissue Proteins/metabolism , Neurofibromin 1/metabolism , Neuromuscular Junction/pathology , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Rats , Rats, Inbred Lew , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism
10.
Stroke ; 49(1): 184-192, 2018 01.
Article in English | MEDLINE | ID: mdl-29212744

ABSTRACT

BACKGROUND AND PURPOSE: Intracerebral hemorrhage (ICH) is a devastating disease without effective treatment. As a key component of the innate immune system, the NOD-like receptor (NLR) family, NLRP3 (pyrin domain-containing protein 3) inflammasome, when activated after ICH, promotes neuroinflammation and brain edema. MCC950 is a potent, selective, small-molecule NLRP3 inhibitor that blocks NLRP3 activation at nanomolar concentrations. Here, we examined the effect of MCC950 on brain injury and inflammation in 2 models of ICH in mice. METHODS: In mice with ICH induced by injection of autologous blood or bacterial collagenase, we determined the therapeutic potential of MCC950 and its mechanisms of neuroprotection. RESULTS: MCC950 reduced IL-1ß (interleukin-1ß) production and attenuated neurodeficits and perihematomal brain edema after ICH induction by injection of either autologous blood or collagenase. In mice with autologous blood-induced ICH, the protection of MCC950 was associated with reduced leukocyte infiltration into the brain and microglial production of IL-6. MCC950 improved blood-brain barrier integrity and diminished cell death. Notably, the protective effect of MCC950 was abolished in mice depleted of either microglia or Gr-1+ myeloid cells. CONCLUSIONS: These results indicate that the NLRP3 inflammasome inhibitor, MCC950, attenuates brain injury and inflammation after ICH. Hence, NLRP3 inflammasome inhibition is a potential therapy for ICH that warrants further investigation.


Subject(s)
Brain Injuries/drug therapy , Cerebral Hemorrhage/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Female , Interleukin-1beta/metabolism , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
11.
J Neuroinflammation ; 14(1): 248, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29246244

ABSTRACT

BACKGROUND: Evidence suggests that brain infiltration of lymphocytes contributes to acute neural injury after cerebral ischemia. However, the spatio-temporal dynamics of brain-infiltrating lymphocytes during the late stage after cerebral ischemia remains unclear. METHODS: C57BL/6 (B6) mice were subjected to sham, photothrombosis, or 60-min transient middle cerebral artery occlusion (MCAO) procedures. Infarct volume, neurodeficits, production of reactive oxygen species (ROS) and inflammatory factors, brain-infiltrating lymphocytes, and their activation as well as pro-inflammatory cytokine IFN-γ production were assessed. Brain-infiltrating lymphocytes were also measured in tissue sections from post-mortem patients after ischemic stroke by immunostaining. RESULTS: In mice subjected to transient MCAO or photothrombotic stroke, we found that lymphocyte infiltration persists in the ischemic brain until at least day 14 after surgery, during which brain infarct volume significantly diminished. These brain-infiltrating lymphocytes express activation marker CD69 and produce proinflammatory cytokines such as IFN-γ, accompanied with a sustained increase of reactive oxygen species (ROS) and inflammatory cytokines release in the brain. In addition, brain-infiltrating lymphocytes were observed in post-mortem brain sections from patients during the late stage of ischemic stroke. CONCLUSION: Our results demonstrate that brain-infiltration of lymphocytes persists after the acute stage of cerebral ischemia, facilitating future advanced studies to reveal the precise role of lymphocytes during late stage of stroke.


Subject(s)
Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Lymphocytes/immunology , Stroke/immunology , Stroke/pathology , Aged , Aged, 80 and over , Animals , Chemotaxis, Leukocyte , Disease Models, Animal , Female , Humans , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL
12.
Proc Natl Acad Sci U S A ; 114(30): E6202-E6211, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696300

ABSTRACT

The nonneural cholinergic system of immune cells is pivotal for the maintenance of immunological homeostasis. Here we demonstrate the expression of choline acetyltransferase (ChAT) and cholinergic enzymes in murine natural killer (NK) cells. The capacity for acetylcholine synthesis by NK cells increased markedly under inflammatory conditions such as experimental autoimmune encephalomyelitis (EAE), in which ChAT expression escalated along with the maturation of NK cells. ChAT+ and ChAT- NK cells displayed distinctive features in terms of cytotoxicity and chemokine/cytokine production. Transfer of ChAT+ NK cells into the cerebral ventricles of CX3CR1-/- mice reduced brain and spinal cord damage after EAE induction, and decreased the numbers of CNS-infiltrating CCR2+Ly6Chi monocytes. ChAT+ NK cells killed CCR2+Ly6Chi monocytes directly via the disruption of tolerance and inhibited the production of proinflammatory cytokines. Interestingly, ChAT+ NK cells and CCR2+Ly6Chi monocytes formed immune synapses; moreover, the impact of ChAT+ NK cells was mediated by α7-nicotinic acetylcholine receptors. Finally, the NK cell cholinergic system up-regulated in response to autoimmune activation in multiple sclerosis, perhaps reflecting the severity of disease. Therefore, this study extends our understanding of the nonneural cholinergic system and the protective immune effect of acetylcholine-producing NK cells in autoimmune diseases.


Subject(s)
Acetylcholine/metabolism , Central Nervous System/pathology , Encephalomyelitis/pathology , Killer Cells, Natural/physiology , Macrophages/physiology , Monocytes/physiology , Animals , Encephalomyelitis/immunology , Humans , Killer Cells, Natural/metabolism , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Non-Neuronal Cholinergic System/immunology , Non-Neuronal Cholinergic System/physiology
13.
J Neuroinflammation ; 14(1): 151, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28754131

ABSTRACT

BACKGROUND: Cerebral ischemia is a leading cause of death and disability with limited treatment options. Although inflammatory and immune responses participate in ischemic brain injury, the molecular regulators of neuroinflammation after ischemia remain to be defined. Translocator protein 18 kDa (TSPO) mainly localized to the mitochondrial outer membrane is predominantly expressed in glia within the central nervous system during inflammatory conditions. This study investigated the effect of a TSPO agonist, etifoxine, on neuroinflammation and brain injury after ischemia/reperfusion. METHODS: We used a mouse model of middle cerebral artery occlusion (MCAO) to examine the therapeutic potential and mechanisms of neuroprotection by etifoxine. RESULTS: TSPO was upregulated in Iba1+ or CD11b+CD45int cells from mice subjected to MCAO and reperfusion. Etifoxine significantly attenuated neurodeficits and infarct volume after MCAO and reperfusion. The attenuation was pronounced in mice subjected to 30, 60, or 90 min MCAO. Etifoxine reduced production of pro-inflammatory factors in the ischemic brain. In addition, etifoxine treatment led to decreased expression of interleukin-1ß, interleukin-6, tumor necrosis factor-α, and inducible nitric oxide synthase by microglia. Notably, the benefit of etifoxine against brain infarction was ablated in mice depleted of microglia using a colony-stimulating factor 1 receptor inhibitor. CONCLUSIONS: These findings indicate that the TSPO agonist, etifoxine, reduces neuroinflammation and brain injury after ischemia/reperfusion. The therapeutic potential of targeting TSPO requires further investigations in ischemic stroke.


Subject(s)
Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Oxazines/therapeutic use , Receptors, GABA/metabolism , Reperfusion Injury/drug therapy , Animals , Brain Edema/drug therapy , Brain Edema/etiology , Brain Infarction/drug therapy , Brain Infarction/etiology , Cytokines/metabolism , Disease Models, Animal , Drug Administration Schedule , Flow Cytometry , Mice , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neurologic Examination , RNA, Messenger , Reperfusion Injury/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
14.
Mult Scler Relat Disord ; 13: 58-66, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28427704

ABSTRACT

BACKGROUND: The spinal cord and brain measurements are rarely investigated in neuromyelitis optica (NMO) patients with and without antibodies to aquaporin-4 (AQP4), directly compared to multiple sclerosis (MS) patients. OBJECTIVES: To investigate magnetic resonance imaging (MRI) features of both brain and spinal cord in NMO patients with and without antibodies to AQP4, compared with MS patients and healthy controls (HC). METHODS: We recruited 55 NMO including 30 AQP4 (+) and 25 AQP4 (-), 25 MS and 25 HC. Brain and spinal cord MRIs were obtained for each participant. Brain lesions (BL), whole brain and deep grey matter volumes (DGMV), white matter diffusion metrics and spinal cord lesions were measured and compared among groups. RESULTS: The incidence of BL was lower in the AQP4 (+) group than in the AQP4 (-) and MS groups (p<0.05). In the AQP4 (+) group, there was a lower incidence of infratentorial lesions (ITL) and higher spinal cord lesions length than in the MS group (p<0.05). The thalamic and hippocampal volumes were smaller in the AQP4 (-) group and MS group than in the HC group (p<0.05). CONCLUSIONS: The NMO patients with AQP4 (-) showed higher prevalence of BL, ITL, and similar spinal cord lesion length, compared to AQP4 (+), and demonstrated deep grey matter atrophy, suggesting an intermediate phenotype between that of typical MS and NMO.


Subject(s)
Aquaporin 4/blood , Brain/diagnostic imaging , Brain/pathology , Neuromyelitis Optica/diagnostic imaging , Neuromyelitis Optica/pathology , Spinal Cord/diagnostic imaging , Spinal Cord/pathology , Adolescent , Adult , Aged , Antibodies , Aquaporin 4/immunology , Female , Gray Matter/diagnostic imaging , Gray Matter/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuromyelitis Optica/blood , Neuromyelitis Optica/immunology , White Matter/diagnostic imaging , White Matter/pathology , Young Adult
15.
J Cereb Blood Flow Metab ; 37(6): 2224-2236, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28273719

ABSTRACT

Brain ischemia elicits microglial activation and microglia survival depend on signaling through colony-stimulating factor 1 receptor (CSF1R). Although depletion of microglia has been linked to worse stroke outcomes, it remains unclear to what extent and by what mechanisms activated microglia influence ischemia-induced inflammation and injury in the brain. Using a mouse model of transient focal cerebral ischemia and reperfusion, we demonstrated that depletion of microglia via administration of the dual CSF1R/c-Kit inhibitor PLX3397 exacerbates neurodeficits and brain infarction. Depletion of microglia augmented the production of inflammatory mediators, leukocyte infiltration, and cell death during brain ischemia. Of note, microglial depletion-induced exacerbation of stroke severity did not solely depend on lymphocytes and monocytes. Importantly, depletion of microglia dramatically augmented the production of inflammatory mediators by astrocytes after brain ischemia . In vitro studies reveal that microglia restricted ischemia-induced astrocyte response and provided neuroprotective effects. Our findings suggest that neuroprotective effects of microglia may result, in part, from its inhibitory action on astrocyte response after ischemia.


Subject(s)
Brain Ischemia/immunology , Brain Ischemia/pathology , Inflammation Mediators/metabolism , Microglia/immunology , Microglia/pathology , Aminopyridines/pharmacology , Animals , Brain Ischemia/diagnostic imaging , Cells, Cultured , Disease Models, Animal , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrroles/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
16.
Proc Natl Acad Sci U S A ; 114(3): E396-E405, 2017 01 17.
Article in English | MEDLINE | ID: mdl-27994144

ABSTRACT

Astrocytes are believed to bridge interactions between infiltrating lymphocytes and neurons during brain ischemia, but the mechanisms for this action are poorly understood. Here we found that interleukin-15 (IL-15) is dramatically up-regulated in astrocytes of postmortem brain tissues from patients with ischemic stroke and in a mouse model of transient focal brain ischemia. We generated a glial fibrillary acidic protein (GFAP) promoter-controlled IL-15-expressing transgenic mouse (GFAP-IL-15tg) line and found enlarged brain infarcts, exacerbated neurodeficits after the induction of brain ischemia. In addition, knockdown of IL-15 in astrocytes attenuated ischemic brain injury. Interestingly, the accumulation of CD8+ T and natural killer (NK) cells was augmented in these GFAP-IL-15tg mice after brain ischemia. Of note, depletion of CD8+ T or NK cells attenuated ischemic brain injury in GFAP-IL-15tg mice. Furthermore, knockdown of the IL-15 receptor α or blockade of cell-to-cell contact diminished the activation and effector function of CD8+ T and NK cells in GFAP-IL-15tg mice, suggesting that astrocytic IL-15 is delivered in trans to target cells. Collectively, these findings indicate that astrocytic IL-15 could aggravate postischemic brain damage via propagation of CD8+ T and NK cell-mediated immunity.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Brain Ischemia/metabolism , Immunity, Cellular/immunology , Interleukin-15/immunology , Interleukin-15/metabolism , Aged, 80 and over , Animals , Astrocytes/immunology , Brain/immunology , Brain/metabolism , Brain Injuries/immunology , Brain Ischemia/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/immunology , Glial Fibrillary Acidic Protein/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic/immunology , Mice, Transgenic/metabolism , Neuroglia/immunology , Neuroglia/metabolism , Neurons/immunology , Neurons/metabolism , Promoter Regions, Genetic/immunology , Stroke/immunology , Stroke/metabolism , Up-Regulation/immunology
17.
FASEB J ; 31(1): 172-179, 2017 01.
Article in English | MEDLINE | ID: mdl-27671228

ABSTRACT

Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons with limited treatment options. Emerging evidence shows that FTY720 protects against neural injury via modulation of the sphingosine-1-phosphate 1 receptor (S1PR1). However, it remains unclear whether FTY720 could influence neurodegeneration in PD. Therefore, the present study was designed to determine the impact of fingolimod (FTY720), a sphingosine-1-phosphate receptor (S1PR) agonist, on 2 mouse models of PD. We found that FTY720 significantly reduced the deficit of motor function, diminished the loss of tyrosine hydroxylase-positive neurons in the substantia nigra, and attenuated the decrease of striatal dopamine and metabolite levels in mice receiving 6-hydroxydopamine (6-OHDA) or rotenone to simulate PD. An S1PR1-selective antagonist, W146, blocked the neuroprotective effects of FTY720. Of note, FTY720 retained the phosphorylation of ERK, together with a decreased expression of cleaved caspase-3 in mice treated with 6-OHDA or rotenone. In vitro studies revealed that FTY720 also attenuated 6-OHDA- or rotenone-induced toxicity in SH-SY5Y cells. These findings suggest the potential of S1PR modulation as a treatment for PD.-Zhao, P., Yang, X., Yang, L., Li, M., Wood, K., Liu, Q., Zhu, X. Neuroprotective effects of fingolimod in mouse models of Parkinson's disease.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Animals , Cell Line, Tumor , Gene Expression Regulation/physiology , Humans , Male , Mice , Neuroblastoma/drug therapy , Oxidopamine/pharmacology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Rotenone/toxicity , Sphingosine-1-Phosphate Receptors
18.
J Cereb Blood Flow Metab ; 37(7): 2383-2395, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27596835

ABSTRACT

Microglia are the first responders to intracerebral hemorrhage, but their precise role in intracerebral hemorrhage remains to be defined. Microglia are the only type of brain cells expressing the colony-stimulating factor 1 receptor, a key regulator for myeloid lineage cells. Here, we determined the effects of a colony-stimulating factor 1 receptor inhibitor (PLX3397) on microglia and the outcome in the context of experimental mouse intracerebral hemorrhage. We show that PLX3397 effectively depleted microglia, and the depletion of microglia was sustained after intracerebral hemorrhage. Importantly, colony-stimulating factor 1 receptor inhibition attenuated neurodeficits and brain edema in two experimental models of intracerebral hemorrhage induced by injection of collagenase or autologous blood. The benefit of colony-stimulating factor 1 receptor inhibition was associated with reduced leukocyte infiltration in the brain and improved blood-brain barrier integrity after intracerebral hemorrhage, and each observation was independent of lesion size or hematoma volume. These results demonstrate that suppression of colony-stimulating factor 1 receptor signaling ablates microglia and confers protection after intracerebral hemorrhage.


Subject(s)
Aminopyridines/pharmacology , Brain/metabolism , Cerebral Hemorrhage/metabolism , Microglia/metabolism , Pyrroles/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Brain/diagnostic imaging , Brain/pathology , Brain Edema/immunology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/prevention & control , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Cytokines/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Neuroimaging , Species Specificity
19.
J Immunol ; 197(9): 3471-3480, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27671112

ABSTRACT

A major hurdle for effective stem cell therapy is ongoing inflammation in the target organ. Reconditioning the lesion microenvironment may be an effective way to promote stem cell therapy. In this study, we showed that engineered neural stem cells (NSCs) with complement factor H-related protein 1, a complement inhibitor protein, can attenuate inflammatory infiltration and immune-mediated damage of astrocytes, an important pathogenic progress in patients with neuromyelitis optica spectrum disorders. Furthermore, we demonstrated that transplantation of the complement factor H-related protein 1-modified NSCs effectively blocked the complement activation cascade and inhibited formation of the membrane attack complex, thus contributing to the protection of endogenous and transplanted NSC-differentiated astrocytes. Therefore, manipulation of the lesion microenvironment contributes to a more effective cell replacement therapeutic strategy for autoimmune diseases of the CNS.


Subject(s)
Astrocytes/physiology , Complement C3b Inactivator Proteins/metabolism , Neural Stem Cells/physiology , Neuromyelitis Optica/immunology , Neuroprotection , Stem Cell Transplantation , Adult , Animals , Aquaporin 4/immunology , Autoantibodies/metabolism , Autoantigens/immunology , Cells, Cultured , Complement Activation , Complement C3b Inactivator Proteins/genetics , Disease Models, Animal , Female , Genetic Therapy , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Neural Stem Cells/transplantation , Neuromyelitis Optica/therapy , Young Adult
20.
J Immunol ; 197(7): 2610-7, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27543617

ABSTRACT

Myasthenia gravis (MG) is a chronic humoral immunity-mediated autoimmune disorder of the neuromuscular junction characterized by muscle weakness. Follicular helper T (Tfh) cells may be the key Th cell subset that promotes MG development, as their major function is helping B cell activation and Ab production. Aberrance of thymus-derived Tfh cells might be implicated in autoimmune diseases including MG; just how circulating Tfh cells, especially those from patients with a normal thymus, contribute to MG pathogenesis remains to be uncovered. In this article, we characterize a population of circulating CD4(+)CXCR5(+)PD-1(+) Tfh cells in ocular and generalized MG patients without thymic abnormalities and demonstrate that the circulating Tfh cells are significantly enriched in generalized MG patients but not in ocular MG patients compared with healthy subjects, whereas a proportion of follicular regulatory T cells decreased in MG patients. In addition, the frequency of plasma cells and B cells was higher and the serum levels of IL-6/IL-21 were also elevated in these MG patients. The activated Tfh1 and Tfh17 in Tfh cells are the major source for IL-21 production in MG patients. A strong correlation between Tfh cells and the plasma cell frequency and anti-acetylcholine receptor Ab titers was evident in generalized MG patients. In particular, we found that Tfh cells derived from MG patients promoted B cells to produce Abs in an IL-21 signaling-dependent manner. Collectively, our results suggest that circulating Tfh cells may act on autoreactive B cells and thus contribute to the development of MG in patients without thymic abnormalities.


Subject(s)
B-Lymphocytes/immunology , Myasthenia Gravis/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Myasthenia Gravis/pathology , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...