Recruitment of Regulatory T Cells with rCCL17 Promotes M2 Microglia/Macrophage Polarization Through TGFβ/TGFβR/Smad2/3 Pathway in a Mouse Model of Intracerebral Hemorrhage

Author(s):  
Shuixiang Deng ◽  
Peng Jin ◽  
Shengpeng Liu ◽  
Lei Huang ◽  
Prativa Sherchan ◽  
...  

Abstract Background Intracerebral hemorrhage (ICH) is a devastating neurological disease with high mortality and morbidity. The microglia activation and peripheral inflammatory cells infiltration play an important role in the ICH prognosis. Previous studies have demonstrated that regulatory T cells (Tregs) ameliorated neuroinflammation following experimental ICH. However, the specific molecular mechanism underlying such effects of Tregs remains unclear. In the present study, our aims were to examine the effect of Tregs recruitment induced by recombinant CC chemokine ligand 17(rCCL17) in an intrastriatal autologous blood mouse model of ICH and to determine whether the TGF-β/TGF-βR/Smad2/3 pathway was involved in Tregs promoted M2 microglia/macrophage polarization. Methods A total of 404 adult CD1 mice (male, eight-week-old) were subject to sham surgery or autologous blood injection of ICH. A CD25-specific mouse antibody or isotype control mAb was injected intraperitoneally 48h prior to ICH induction to deplete Tregs. Recombinant CCL17 (rCCL17), a C-C chemokine receptor 4 (CCR4), was delivered intranasally at 1 h post-ICH. SB431542, a specific inhibitor of TGF-β was administered intraperitoneally 1 h before ICH induction. Post-ICH assessments included neuro-behavior evaluation, brain edema, hematoma volume, hemoglobin content, western blotting, double immunofluorescence staining and immunohistochemistry. Results Endogenous brain expressions of CCL17 and Tregs marker Foxp3 as well as the number of Tregs in the perihematomal region were increased following ICH. The Tregs deletion by a CD25 antibody aggravated short-term neurological deficits and brain edema, increased the level of inflammatory cytokines and peripheral inflammatory cells infiltration, exacerbated hematoma expansion and increased M1phenotypes of microglia/macrophage in ICH mice. The rCCL17 treatment increased the number of Tregs in the brain, reduced hematoma expansion and brain edema, promoted microglia/macrophage polarization toward M2 phenotypes. Moreover, the expressions of brain TGF-β/phosphorylated-Smad2/3 were increased. The neuroprotective effects of rCCL17 were abolished by co-administration of the selective TGF-β inhibitor SB431542. Conclusions Our study demonstrated rCCL17 recruited of Tregs to brain in the autologous blood injection model of ICH. Tregs promoted microglia/macrophages polarization toward M2 phenotype and alleviation early brain injury, at least in part, through the TGFβ/TGFβR/Smad2/3 signaling pathway in ICH mice. Thus, rCCL17-mediated Tregs recruitment may provide a promising therapeutic strategy to reduce early brain injury after ICH.

2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Shuixiang Deng ◽  
Peng Jin ◽  
Prativa Sherchan ◽  
Shengpeng Liu ◽  
Yuhui Cui ◽  
...  

Abstract Background Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance after ICH. However, it is unclear whether CCR4 activation can ameliorate neuroinflammation and apoptosis of neurons following ICH. The aim of the present study was to examine the effects of recombinant CCL17 (rCCL17)-dependent CCR4 activation on neuroinflammation and neuronal apoptosis in an intrastriatal autologous blood injection ICH model, and to determine whether the PI3K/AKT/Foxo1 signaling pathway was involved. Methods Two hundred twenty-six adult (8-week-old) male CD1 mice were randomly assigned to sham and ICH surgery groups. An intrastriatal autologous blood injection ICH model was used. rCCL17, a CCR4 ligand, was delivered by intranasal administration at 1 h, 3 h, and 6 h post-ICH. CCL17 antibody was administrated by intraventricular injection at 1 h post-ICH. C021, a specific inhibitor of CCR4 and GDC0068, an AKT inhibitor were delivered intraperitoneally 1 h prior to ICH induction. Brain edema, neurobehavioral assessments, western blotting, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunofluorescence staining were conducted. Results Endogenous expression of CCL17 and CCR4 were increased following ICH, peaking at 5 days post-induction. CCR4 was found to co-localize with microglia, neurons, and astrocytes. rCCL17 treatment decreased brain water content, attenuated short- and long-term neurological deficits, deceased activation of microglia/macrophages and infiltration of neutrophils, and inhibited neuronal apoptosis in the perihematomal region post-ICH. Moreover, rCCL17 treatment post-ICH significantly increased the expression of CCR4, PI3K, phosphorylated AKT, and Bcl-2, while Foxo1, IL-1β, TNF-α, and Bax expression were decreased. The neuroprotective effects of rCCL17 were reversed with the administration of C021 or GDC0068. Conclusions rCCL17-dependent CCR4 activation ameliorated neurological deficits, reduced brain edema, and ameliorated neuroinflammation and neuronal apoptosis, at least in part, through the PI3K/AKT/Foxo1 signaling pathway after ICH. Thus, activation of CCR4 may provide a promising therapeutic approach for the early management of ICH.


2012 ◽  
Vol 2012 ◽  
pp. 1-6 ◽  
Author(s):  
Nicola Massy-Westropp ◽  
Stuart Simmonds ◽  
Suzanne Caragianis ◽  
Andrew Potter

Purpose. This study explored the effect of autologous blood injection (with ultrasound guidance) to the elbows of patients who had radiologically assessed degeneration of the origin of extensor carpi radialis brevis and failed cortisone injection/s to the lateral epicondylitis.Methods. This prospective longitudinal series involved preinjection assessment of pain, grip strength, and function, using the patient-rated tennis elbow evaluation. Patients were injected with blood from the contralateral limb and then wore a customised wrist support for five days, after which they commenced a stretching, strengthening, and massage programme with an occupational therapist. These patients were assessed after six months and then finally between 18 months and five years after injection, using the patient-rated tennis elbow evaluation.Results. Thirty-eight of 40 patients completed the study, showing significant improvement in pain; the worst pain decreased by two to five points out of a 10-point visual analogue for pain. Self-perceived function improved by 11–25 points out of 100. Women showed significant increase in grip, but men did not.Conclusions. Autologous blood injection improved pain and function in a worker’s compensation cohort of patients with chronic lateral epicondylitis, who had not had relief with cortisone injection.


2001 ◽  
Vol 132 (1) ◽  
pp. 36-40 ◽  
Author(s):  
Alan Burnstein ◽  
Darrell WuDunn ◽  
Yoko Ishii ◽  
Christian Jonescu-Cuypers ◽  
Louis B Cantor

1997 ◽  
Vol 28 (10) ◽  
pp. 866-868
Author(s):  
Ramesh S Ayyala ◽  
Robert C Urban ◽  
Mina S Krishnamurthy ◽  
David J Mendelblatt

Stroke ◽  
2017 ◽  
Vol 48 (suppl_1) ◽  
Author(s):  
Arthur F Steinschneider ◽  
Che-Feng Chang ◽  
Michael H Askenase ◽  
Youxi Ai ◽  
Lauren H Sansing

Introduction: After intracerebral hemorrhage (ICH), erythrocytes contribute to secondary injury by releasing toxic hemoproteins. Our lab has previously shown that blood derived macrophages play an important role in ICH clearance but mechanisms of phagocytosis by human macrophages are unknown. This study aims to quantify eryptotic (phosphatidylserine (PtdSer)-expressing) red blood cells (RBCs) in an in vivo model of ICH, and to investigate the mechanisms that play a role in autologous eryptotic phagocytosis by human monocyte derived macrophages (huMDMs). Methods: ICH was induced in mice by autologous blood injection. The mice were sacrificed at 1 day after ICH. The brains were separated into hemispheres and digested into a single cell suspension for analysis by flow cytometry. Cells were stained with antibodies to cell surface markers and annexin V to quantify externalized PtdSer expression. Human monocytes were cultured with M-CSF for 7 days to generate huMDMs. Autologous RBCs were heat shocked (HS) to induce eryptosis. The huMDMs were cocultured with HS RBCs, HS RBCs treated with annexin V, or control RBCs. After 1 hour of coculture, the huMDMs were washed, stained and erythrophagocytosis quantified by microscopy. Results: The proportion of cells that externalized PtdSer increased by almost 20 fold at day 1 after ICH. Control brains mixed with fresh RBCs and subjected to tissue prep did not show PtdSer expression, ensuring that the PtdSer expression detected was induced in vivo (Fig A). HS RBCs increased PtdSer expression and were efficiently phagocytosed by huMDMs. Treatment of HS RBCs with annexin V to antagonize PtdSer-receptor interactions decreased RBC phagocytosis to levels comparable to control RBCs (Figs B and C). Conclusions: In vivo after ICH, erythrocytes externalize PtdSer, a cue to be engulfed by macrophages. Human macrophages phagocytose RBCs in a PtdSer-dependent mechanism. These findings highlight potential targets to enhance ICH clearance.


Sign in / Sign up

Export Citation Format

Share Document