Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Eur J Neurosci ; 59(2): 192-207, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38145884

ABSTRACT

Skeletal muscle is striated muscle that moves autonomously and is innervated by peripheral nerves. Peripheral nerve injury is very common in clinical treatment. However, the commonly used treatment methods often focus on the regeneration of the injured nerve but overlook the pathological changes in the injured skeletal muscle. Acupuncture, as the main treatment for denervated skeletal muscle atrophy, is used extensively in clinical practice. In the present study, a mouse model of lower limb sciatic nerve detachment was constructed and treated with electroacupuncture Stomach 36 to observe the atrophy of lower limb skeletal muscle and changes in skeletal muscle fibre types before and after electroacupuncture Stomach 36 treatment. Mice with skeletal muscle denervation showed a decrease in the proportion of IIa muscle fibres and an increase in the proportion of IIb muscle fibres, after electroacupuncture Stomach 36. The changes were reversed by specific activators of p38 MAPK, which increased IIa myofibre ratio. The results suggest that electroacupuncture Stomach 36 can reverse the change of muscle fibre type from IIb to IIa after denervation of skeletal muscle by inhibiting p38 MAPK. The results provide an important theoretical basis for the treatment of clinical peripheral nerve injury diseases with electroacupuncture, in addition to novel insights that could facilitate the study of pathological changes of denervated skeletal muscle.


Subject(s)
Electroacupuncture , Peripheral Nerve Injuries , Rats , Mice , Animals , Rats, Sprague-Dawley , Peripheral Nerve Injuries/therapy , Muscle Fibers, Skeletal , Muscle, Skeletal , Sciatic Nerve/injuries , Muscular Atrophy/therapy , p38 Mitogen-Activated Protein Kinases
2.
FASEB J ; 34(10): 13762-13775, 2020 10.
Article in English | MEDLINE | ID: mdl-32808351

ABSTRACT

Cerebral ischemia causes damage to the structure and function of the blood-brain barrier (BBB) and alleviating BBB destruction will be of great significance for the treatment and prognosis of ischemic stroke. Recently, microRNAs have been shown to play a critical role in BBB integrity. However, the potential mechanism by which microRNA-182 (miR-182) affects the BBB in ischemic stroke remains unclear. We demonstrated for the first time that cerebral ischemia leads to a significant progressive increase in miR-182 after pMCAO, and bEnd.3 cells are the primary target cells of miR-182. In miR-182 KD transgenic mice, infarct volume, and BBB permeability were attenuated, and tight junction (TJ) proteins increased. Inhibition of miR-182 with an antagomir reduced OGD-induced apoptosis of bEnd.3 cells and the loss of ZO-1 and Occludin. To further explore the mechanism by which miR-182 regulates BBB integrity, we detected the apoptotic proteins Bcl-2/Bax and demonstrated that mTOR and FOXO1 were the targets of miR-182. Inhibition of mTOR/FOXO1 by rapamycin/AS1842856 decreased the ratio of Bcl-2/Bax and exacerbated TJ protein loss. Taken together, inhibition of miR-182 protects BBB integrity by reducing endothelial cell apoptosis through the mTOR/FOXO1 pathway. Thus, miR-182 may be a potential target for the treatment of BBB disruption during cerebral ischemia.


Subject(s)
Blood-Brain Barrier/metabolism , Infarction, Middle Cerebral Artery/metabolism , MicroRNAs/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line , Cells, Cultured , Down-Regulation , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Infarction, Middle Cerebral Artery/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
3.
Eur J Immunol ; 49(12): 2184-2194, 2019 12.
Article in English | MEDLINE | ID: mdl-31411745

ABSTRACT

MicroRNA 182 is important for the clonal expansion of CD4+ T cells (Th) following IL-2 stimulation and is a potential therapeutic target for autoimmune diseases. In the present study, we investigated the role of microRNA 182 in the differentiation of pro-inflammatory CD4+ T helper cell by overexpressing or silencing microRNA 182 expression both in in vivo and in vitro settings. We report that in the studied Chinese cohort, microRNA 182 is upregulated in patients with relapse and remitting multiple sclerosis (RRMS) and this upregulation is associated with increased IFN-γ producing CD4+ Th1 cells in the circulation. In the murine experimental autoimmune encephalomyelitis (EAE) model, global microRNA 182 overexpression exacerbates clinical symptoms and results in augmented CD4+ IFN-γ+ Th1 and CD4+ IL-17+ Th17 differentiation in vivo. Addition of microRNA 182 mimics in vitro represses both the protein expression and transcriptional activity of hypoxia induced factor 1α (HIF-1α) but increases the level of IFN-γ transcripts in sorted murine CD4+ T cells. Together, our results provide evidence that microRNA 182 may be one of the transitional hubs contribution to regulate Th cells expansion in response to self-antigens and differentiation of antigen specific Th cells during the progression of autoimmune inflammations.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , MicroRNAs/immunology , Multiple Sclerosis/immunology , Th1 Cells/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Multiple Sclerosis/pathology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
4.
Clin Sci (Lond) ; 133(7): 869-884, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30914441

ABSTRACT

Background: Early strut coverage after sirolimus-eluting stent (SES) implantation is associated with the activation of inflammation, but the underlying mechanisms are not completely understood. The present study aimed to identify the relationship between the anti-inflammatory cytokine interleukin (IL) 35 (IL-35) and early strut coverage in vivo and in vitroMethods: We utilized a retrospective study design to measure IL-35 levels in 68 stents from 68 patients with coronary artery disease and recorded serial optical coherence tomography (OCT) images (0 and 3 months) to assess stent endothelialization. The mechanism underlying the regulatory effects of IL-35 on macrophages and human umbilical vein endothelial cells (HUVECs) was also investigated. SESs were surgically implanted into the right common carotid arteries of 200 male New Zealand White rabbits receiving intravenous injections of IL-35 or a placebo.Results: At the 3-month OCT evaluation, complete endothelium coverage was correlated with IL-35 levels. IL-35 induced the activation of an anti-inflammatory M2-like macrophage phenotype by targeting the signal transducer and activators of transcription (STAT)1/4 signalling pathway, and IL-35-treated macrophages induced endothelial proliferation and alleviated endothelial dysfunction. IL-35-treated New Zealand White rabbits with implanted SESs showed lower percentages of cross-sections with an uncovered strut, elevated mean neointimal hyperplasia (NIH) thickness, and inhibited inflammatory responses.Conclusions: We investigated the effect of IL-35 expression on early stent endothelialization in vivo and in vitro and identified a crucial role for IL-35 in inducing the activation of an anti-inflammatory M2-like macrophage phenotype. The present study highlights a new therapeutic strategy for early stent endothelialization.


Subject(s)
Cell Proliferation , Coronary Artery Disease/therapy , Coronary Vessels/metabolism , Drug-Eluting Stents , Endothelial Cells/metabolism , Interleukins/blood , Macrophage Activation , Macrophages/metabolism , Percutaneous Coronary Intervention/instrumentation , Aged , Animals , Biomarkers/blood , Cell Proliferation/drug effects , Cells, Cultured , Coronary Artery Disease/blood , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/pathology , Coronary Vessels/diagnostic imaging , Coronary Vessels/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Humans , Interleukins/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Male , Middle Aged , Models, Animal , Percutaneous Coronary Intervention/adverse effects , Rabbits , Retrospective Studies , Time Factors , Tomography, Optical Coherence , Treatment Outcome , Up-Regulation
5.
J Immunol ; 198(8): 3245-3254, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28275140

ABSTRACT

Fungal infections (e.g., Candida albicans) can manifest as serious medical illnesses, especially in the elderly and immune-compromised hosts. T cells are important for Candida control. Whether and how B cells are involved in antifungal immunity has been less clear. Although patients with agammaglobulinemia exhibit normal antifungal immunity, increased fungal infections are reported following B cell-depleting therapy, together pointing to Ab-independent roles of B cells in controlling such infections. To test how human B cells may contribute to fungal-associated human T cell responses, we developed a novel Ag-specific human T cell/B cell in vitro coculture system and found that human B cells could induce C. albicans-associated, MHC class II-restricted responses of naive T cells. Activated B cells significantly enhanced C. albicans-mediated Th1 and Th17 T cell responses, which were both strongly induced by CD80/CD86 costimulation. IL-6+GM-CSF+ B cells were the major responding B cell subpopulation to C. albicans and provided efficient costimulatory signals to the T cells. In vivo B cell depletion in humans resulted in reduced C. albicans-associated T responses. Of note, the decreased Th17, but not Th1, responses could be reversed by soluble factors from B cells prior to depletion, in an IL-6-dependent manner. Taken together, our results implicate an Ab-independent cytokine-defined B cell role in human antifungal T cell responses. These findings may be particularly relevant given the prospects of chronic B cell depletion therapy use in lymphoma and autoimmune disease, as patients age and are exposed to serial combination therapies.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Mycoses/immunology , Candida albicans/immunology , Coculture Techniques , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans
6.
J Immunol ; 198(2): 691-698, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27974457

ABSTRACT

The therapeutic mode of action of dimethyl fumarate (DMF), approved for treating patients with relapsing-remitting multiple sclerosis, is not fully understood. Recently, we and others demonstrated that Ab-independent functions of distinct B cell subsets are important in mediating multiple sclerosis (MS) relapsing disease activity. Our objective was to test whether and how DMF influences both the phenotype and functional responses of disease-implicated B cell subsets in patients with MS. High-quality PBMC were obtained from relapsing-remitting MS patients prior to and serially after initiation of DMF treatment. Multiparametric flow cytometry was used to monitor the phenotype and functional response-profiles of distinct B cell subsets. Total B cell counts decreased following DMF treatment, largely reflecting losses of circulating mature/differentiated (but not of immature transitional) B cells. Within the mature B cell pool, DMF had a greater impact on memory than naive B cells. In keeping with these in vivo effects, DMF treatment in vitro remarkably diminished mature (but not transitional B cell) survival, mediated by inducing apoptotic cell death. Although DMF treatment (both in vivo and in vitro) minimally impacted B cell IL-10 expression, it strongly reduced B cell expression of GM-CSF, IL-6, and TNF-α, resulting in a significant anti-inflammatory shift of B cell response profiles. The DMF-mediated decrease in B cell proinflammatory cytokine responses was further associated with reduced phosphorylation of STAT5/6 and NF-κB in surviving B cells. Together, these data implicate novel mechanisms by which DMF may modulate MS disease activity through shifting the balance between pro- and anti-inflammatory B cell responses.


Subject(s)
B-Lymphocyte Subsets/drug effects , Dimethyl Fumarate/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/immunology , Adult , B-Lymphocyte Subsets/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Young Adult
7.
Genomics ; 110(6): 337-346, 2018 11.
Article in English | MEDLINE | ID: mdl-29366861

ABSTRACT

Long non-coding RNAs have the potential to regulate immune responses. Their impact on multiple sclerosis has remained elusive. For illustrating their roles in experimental autoimmune encephalomyelitis (EAE) pathogenesis, we investigated the differential expression of lncRNAs and mRNAs in CD4+Th cells obtained from myelin oligodendrocytic glycoprotein35-55(MOG35-55)-induced EAE and complete Freund's adjuvant (CFA) controls. We observed differential expression of 1112 lncRNAs and 519 mRNAs in CD4+Th cells. The functional network showed lncRNAs had the capacity to modulate EAE pathogenesis via regulating many known EAE regulators such as Ptpn6. Predicting the function of lncRNAs demonstrated that dysregulated lncRNAs were closely associated with the development of EAE. These dysregulated lncRNAs may have function in EAE and they could be novel biomarkers and therapeutic targets of EAE. However, the precise mechanisms and biological functions of these specific lncRNAs in EAE pathogenesis require further study.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Gene Expression Profiling , Mice , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , T-Lymphocytes, Helper-Inducer/drug effects
8.
Cell Physiol Biochem ; 45(1): 258-266, 2018.
Article in English | MEDLINE | ID: mdl-29402798

ABSTRACT

BACKGROUND/AIMS: Ischemic stroke is a major cause of disability and mortality worldwide, while effective restorative treatments are limited at present. Stem cell transplantation holds therapeutic potential for ischemic vascular diseases and may provide an opportunity for neural regeneration. Dental pulp stem cells (DPSCs) origin from neural crest and have neuro-ectodermal features including proliferation and multilineage differentiation potentials. METHODS: The rat model of middle cerebral artery occlusion (MCAO) was used to evaluate whether intravenous administration of DPSCs can reduce infarct size and to estimate the migration and trans-differentiation into neuron-like cells in focal cerebral ischemia models. Brain tissues were collected at 4 weeks following cell transplantation and analyzed with immunofluorescence, immunohistochemistry and real-time polymerase chain reaction (RT-PCR) methods. RESULTS: Intravenously administration of rat-derived DPSCs were found to migrate into the boundary of ischemic areas and expressed neural specific markers, reducing infarct volume and cerebral edema. CONCLUSIONS: These results suggest that DPSCs treatment may serve as a potential therapy for clinical stroke patients in the future.


Subject(s)
Biomarkers/metabolism , Brain Ischemia/therapy , Stem Cell Transplantation , Animals , Brain/pathology , Brain Ischemia/pathology , Cells, Cultured , Dental Pulp/cytology , Disease Models, Animal , Doublecortin Domain Proteins , Male , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Nestin/metabolism , Neuropeptides/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/metabolism , Tubulin/metabolism
9.
Immunol Invest ; 47(7): 712-724, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29944018

ABSTRACT

Interleukin-9 (IL-9) is a multifunctional cytokine involved in protective immunity or immunopathology depending on the microenvironment and specific disease settings. Our early study determined that IL-9 and Th9 cells participate in and promote the progression of experimental autoimmune myasthenia gravis (EAMG). The data from this study showed that exogenous recombinant rat IL-9 (rrIL-9) acted as an IL-9 receptor antagonist, reduced the incidence of EAMG in rats, alleviated the severity of the disease, and reduced the anti-acetylcholine receptor (AChR) IgG antibody levels by altering the Th-subset distribution. These data suggest that administration of rrIL-9 may provide a novel therapeutic strategy against MG or related autoimmune diseases. Abbreviations: 2-Mercaptoethanol (2-ME); antibodies (Abs); ?-bungarotoxin (?-BTX); acetylcholine receptor (AChR); airway hyper-reactivity (AHR); allophycocyanin-conjugated (APC); antigen presenting cells (APCs); complete Freund's adjuvant (CFA); Cyanine dye 3 (Cy3); dendritic cells (DCs); experimental autoimmune encephalomyelitis (EAE); experimental autoimmune myasthenia gravis (EAMG); flow cytometry (FACS); fetal bovine serum (FBS); fetal calf serum (FCS); Fluorescein isothiocyanate (FITC); gamma chain (?c); intraperitoneally (i.p.); Incomplete Freund's adjuvant (IFA); interferon (IFN); immunoglobulin (Ig); Interleukin (IL); Janus kinase (JAK); myasthenia gravis (MG); Mononuclear cells (MNC); neuromuscular junctions (NMJ); optical density (OD); ovalbumin (OVA); phosphate-buffered saline (PBS); phycoerythrin (PE); Peridinin chlorophyll protein complex (Percp); Rat AChR ? subunit (R-AChR97-116); Recombinant Rat (rr); room temperature (RT); signal transducer and activator of transcription (STAT); T helper cells (Th).


Subject(s)
Immunotherapy/methods , Interleukin-9/immunology , Myasthenia Gravis, Autoimmune, Experimental/therapy , Myasthenia Gravis/therapy , Recombinant Proteins/immunology , Animals , Autoantibodies/blood , Autoantigens/immunology , Female , Humans , Interleukin-9/therapeutic use , Myasthenia Gravis/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Peptides/immunology , Rats , Rats, Inbred Lew , Receptors, Cholinergic/immunology , Receptors, Interleukin-9/antagonists & inhibitors , Recombinant Proteins/therapeutic use
10.
Mol Cell Neurosci ; 83: 65-73, 2017 09.
Article in English | MEDLINE | ID: mdl-28684360

ABSTRACT

Regulator of G protein signaling 5 (RGS5) acts as a GTPase-activating protein (GAP) for the Gαi subunit and negatively regulates G protein-coupled receptor signaling. However, its presence and function in postmitotic differentiated primary neurons remains largely uncharacterized. During neural development, sonic hedgehog (Shh) signaling is involved in cell signaling pathways via Gαi activity. In particular, Shh signaling is essential for embryonic neural tube patterning, which has been implicated in neuronal polarization involving neurite outgrowth. Here, we examined whether RGS5 regulates Shh signaling in neurons. RGS5 transcripts were found to be expressed in cortical neurons and their expression gradually declined in a time-dependent manner in culture system. When an adenovirus expressing RGS5 was introduced into an in vitro cell culture model of cortical neurons, RGS5 overexpression significantly reduced neurite outgrowth and FM4-64 uptake, while cAMP-PKA signaling was also affected. These findings suggest that RGS5 inhibits Shh function during neurite outgrowth and the presynaptic terminals of primary cortical neurons mature via modulation of cAMP.


Subject(s)
Hedgehog Proteins/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Signal Transduction , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Neuronal Outgrowth , Neurons/cytology , RGS Proteins/genetics
11.
EMBO J ; 32(18): 2477-90, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-23942232

ABSTRACT

In rheumatoid arthritis (RA), macrophage is one of the major sources of inflammatory mediators. Macrophages produce inflammatory cytokines through toll-like receptor (TLR)-mediated signalling during RA. Herein, we studied macrophages from the synovial fluid of RA patients and observed a significant increase in activation of inositol-requiring enzyme 1α (IRE1α), a primary unfolded protein response (UPR) transducer. Myeloid-specific deletion of the IRE1α gene protected mice from inflammatory arthritis, and treatment with the IRE1α-specific inhibitor 4U8C attenuated joint inflammation in mice. IRE1α was required for optimal production of pro-inflammatory cytokines as evidenced by impaired TLR-induced cytokine production in IRE1α-null macrophages and neutrophils. Further analyses demonstrated that tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) plays a key role in TLR-mediated IRE1α activation by catalysing IRE1α ubiquitination and blocking the recruitment of protein phosphatase 2A (PP2A), a phosphatase that inhibits IRE1α phosphorylation. In summary, we discovered a novel regulatory axis through TRAF6-mediated IRE1α ubiquitination in regulating TLR-induced IRE1α activation in pro-inflammatory cytokine production, and demonstrated that IRE1α is a potential therapeutic target for inflammatory arthritis.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Cytokines/metabolism , Endoribonucleases/metabolism , Enzyme Activation/physiology , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptors/metabolism , Unfolded Protein Response/physiology , Animals , Blotting, Western , Cell Line , Drug Delivery Systems , Endoribonucleases/antagonists & inhibitors , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Immunoprecipitation , Macrophages/cytology , Macrophages/metabolism , Mice , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Real-Time Polymerase Chain Reaction , Synovial Fluid/cytology , TNF Receptor-Associated Factor 6/pharmacology
12.
Clin Immunol ; 173: 109-116, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27664932

ABSTRACT

MicroRNA 182 has been found to have a distinct contribution in the clonal expansion of activated- and functioning of specialized-helper T cells. In this study we knocked down microRNA 182 in vivo and induced experimental autoimmune encephalomyelitis (EAE) to determine the influences of microRNA 182 in the Treg cells functional specialization through Foxo1 dependent pathway in the peripheral lymphoid organs. Down-regulation of microRNA 182 significantly increased the proportions of Foxp3+ T cells in the peripheral lymph nodes and spleen. In vivo study verified a positive correlation between microRNA 182 levels and symptom severity of EAE, and a negative correlation between microRNA 182 and the transcriptional factor Foxp3. In vitro polarization study also confirmed the contribution of Foxo1 in microRNA 182 mediated down-regulation of Foxp3+ T cells. Together, our results provide evidence that during the development of EAE, microRNA 182 repressed Treg cells differentiation through the Foxo1 dependent pathway.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Box Protein O1/immunology , MicroRNAs/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Female , Lymph Nodes/cytology , Mice, Inbred C57BL , Spleen/cytology , T-Lymphocytes, Regulatory/physiology
13.
Cell Physiol Biochem ; 40(5): 1117-1128, 2016.
Article in English | MEDLINE | ID: mdl-27960154

ABSTRACT

BACKGROUND/AIMS: Mesenchymal stem cell (MSC) transplantation has emerged as an option for the treatment of chronic hepatic cirrhosis, while its therapeutic efficacy could be improved. The bcl-2 gene is anti-apoptotic and can help cell survival and proliferation. Therefore, we explored whether transplanted MSCs with enhanced bcl-2 expression may be beneficial in the treatment of experimental cirrhosis in rats. METHODS: MSCs were isolated from rat bone marrow, expanded in vitro and transfected with adeno-associated virus (AAV) engineered the bcl-2 gene (AAV-bcl-2). Rats with cirrhosis induced by carbon tetrachloride (CCl4) were treated with AAV-bcl-2 infected BMSCs-AAV-bcl-2, with the cells traced in vivo post transplantation. Liver pathology and function were evaluated 7, 14, 21, and 28 days post transplantation, respectively. RESULTS: On day 7 post transplantation, the infused AAV-bcl-2 had integrated into the hepatocyte-like cells (HLCs) that expressed albumin (ALB), Cytokeratin 18 (CK18), and hepatocytes nuclear factor 4a (HNF4a). On day 28 post transplantation, rats in the cirrhosis + BMSCs-AAV-bcl-2 group showed the most dense HLCs, highest mRNA and protein levels of ALB, CK18, and HNF4a, compared to the other groups. Their liver function recovered most rapidly in 4 week observation, while histological sign of cirrhosis remained at the end of this period. CONCLUSION: BMSCs over expressing bcl-2 gene showed better survival, and enhanced the differentiation into hepatocytes-like cells, and appeared to promote the recovery of liver function in rats with experimental cirrhosis.


Subject(s)
Liver Cirrhosis, Experimental/therapy , Liver Regeneration , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Antigens/metabolism , Biomarkers/blood , Carbon Tetrachloride , Cell Shape , Disease Models, Animal , Gene Expression , Genes, Reporter , Hepatocytes/metabolism , Liver Cirrhosis, Experimental/blood , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Male , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reproducibility of Results
15.
Cell Physiol Biochem ; 35(4): 1454-66, 2015.
Article in English | MEDLINE | ID: mdl-25791376

ABSTRACT

BACKGROUND: The accumulation of cytokines in the plasma after trauma can induce myocyte apoptosis. We aimed to identify which cytokine(s) present in the plasma responsible for myocyte apoptosis, and delineated the signal transduction mechanism in rats subjected to surgical trauma. METHODS: Rats were randomized into two groups: control and trauma groups, which was divided into five subgroups: posttraumatic 0, 3, 6, 12, and 24 h subgroups. Cardiomyocytes isolated from traumatized rats were incubated with one of the factors for 12 h (normal plasma; Cytomix; TNF-α; IL-1ß; IFN-γ; trauma plasma; anti-TNF-α antibody; SB203580). Myocyte apoptosis, cytokine levels, and MAPKs activation, as the primary experimental outcomes, were measured by TUNEL, flow cytometry, ELISA and Western blot, respectively. RESULTS: Myocyte apoptosis was induced by surgical trauma during the early stage after trauma. Accompanying this change, plasma TNF-α, IL-1ß, and IFN-γ levels were elevated in traumatized rats. Incubation of traumatized cardiomyocytes with cytomix or TNF-α alone induced myocyte apoptosis, and increased the activation of p38 and ERK1/2. Myocyte apoptosis and p38 activation were elevated in traumatized cardiomyocytes with trauma plasma, and these increases were partly abolished by anti-TNF-α antibody or SB203580. CONCLUSION: Our study demonstrated that there exists the TNF-α-mediated-p38-dependent signaling pathway that contributed to posttraumatic myocyte apoptosis of rats undergoing surgical trauma.


Subject(s)
Apoptosis/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Wounds and Injuries/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies/immunology , Blotting, Western , Caspases/metabolism , Cells, Cultured , Cytokines/analysis , Enzyme-Linked Immunosorbent Assay , Imidazoles/pharmacology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/immunology , Wounds and Injuries/etiology , Wounds and Injuries/metabolism
16.
Dig Dis Sci ; 60(5): 1325-34, 2015 May.
Article in English | MEDLINE | ID: mdl-25445161

ABSTRACT

BACKGROUND: Cirrhosis, or liver fibrosis, which is mainly triggered by cirrhosis fat-storing cells (CFSCs) activation, has traditionally been considered an irreversible disease. However, recent observations indicate that even advanced fibrosis is still reversible by removing the causative agents. Anti-fibrotic effects of bone marrow-derived stromal cells (BMSCs) have been demonstrated by inhibiting CFSCs via cytokines secretion; however, the mechanisms are still unclear. AIMS: The purpose of this study was to explore the underlying mechanisms by which BMSCs modulate the function of activated CFSCs. METHODS: After the co-culture of CFSCs with BMSCs supernatants with or without the addition of recombinant rat adrenomedullin (AM)/AM-specific siRNA, western blot analysis was mainly used to detect the differences of relative protein expression on CFSCs. RESULTS: BMSC-secreted adrenomedullin (AM) effectively inhibited the proliferation and activation of CFSCs by suppressing the expression of Ang II and its binding receptor, AT1, which resulted in a reduction of p47-phox formation. CONCLUSIONS: Our data suggested that BMSCs inhibited CFSC activation in vitro via the AM-Ang II-p47-phox signaling pathway, and since CFSC activation is an essential part of hepatic fibrosis process, this inhibition by BMSCs implies us new insights into the potential treatment of hepatic fibrosis via BMSCs.


Subject(s)
Adrenomedullin/metabolism , Hepatic Stellate Cells/metabolism , Lipid Metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Mesenchymal Stem Cells/metabolism , Paracrine Communication , Actins/metabolism , Adrenomedullin/genetics , Angiotensin II/metabolism , Animals , Cell Proliferation , Cells, Cultured , Coculture Techniques , Collagen Type I/metabolism , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , NADPH Oxidases/metabolism , RNA Interference , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transfection
17.
Immunology ; 143(3): 396-405, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24850614

ABSTRACT

Interleukin-9 (IL-9) was initially thought to be a type 2 T helper (Th2)-associated cytokine involved in the regulation of autoimmune responses by affecting multiple cell types. However, it was recently shown that IL-9-producing CD4+ T cells represent a discrete subset of Th cells, designated Th9 cells. Although Th9 cells have been shown to be important in many diseases, their roles in myasthenia gravis (MG) are unclear. The aim of this study was to determine whether IL-9 and Th9 cells promote the progression of experimental autoimmune myasthenia gravis (EAMG). The results showed that the percentage of Th9 cells changed during the progression of EAMG, accompanied by an up-regulation of IL-9. Blocking IL-9 activity with antibodies against IL-9 inhibited EAMG-associated pathology in rats and reduced serum anti-acetylcholine receptor IgG levels. Neutralization of IL-9 altered the Th subset distribution in EAMG, reducing the number of Th1 cells and increasing the number of regulatory T cells. Administration of an anti-IL-9 antibody may represent an effective therapeutic strategy for MG-associated pathologies or other T-cell- or B-cell-mediated autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Immunity, Humoral , Interleukin-9/antagonists & inhibitors , Myasthenia Gravis, Autoimmune, Experimental/immunology , T-Lymphocyte Subsets/immunology , Animals , Disease Models, Animal , Disease Progression , Female , Interleukin-9/metabolism , Rats , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
18.
Clin Immunol ; 152(1-2): 90-100, 2014.
Article in English | MEDLINE | ID: mdl-24650778

ABSTRACT

In this study, the capacity for t-PA to affect T cell-brain microvascular endothelial cell adhesion by acting as a cytokine was investigated. Following the treatment of a brain-derived endothelial cell line, bEnd.3, with various concentrations of t-PA, adhesion and transwell migration assays were performed. In the presence of t-PA, enhanced adhesion of T cells to bEnd.3 cells was observed. Using western blot analysis, an increase in ICAM-1 expression was detected for both t-PA-treated bEnd.3 cells and bEnd.3 cells treated with a non-enzymatic form of t-PA. In contrast, when LRP1 was blocked using a specific antibody, upregulation of ICAM-1 was inhibited and cAMP-PKA signaling was affected. Furthermore, using an EAE mouse model, administration of t-PA was associated with an increase in ICAM-1 expression by brain endothelial cells. Taken together, these findings suggest that t-PA can induce ICAM-1 expression in brain microvascular endothelial cells, and this may promote the development of EAE.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Multiple Sclerosis/immunology , Tissue Plasminogen Activator/physiology , Animals , Antibodies, Monoclonal/immunology , Brain/blood supply , Cell Adhesion/drug effects , Cell Line, Transformed , Cell Movement/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Intercellular Adhesion Molecule-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Receptors, LDL/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tissue Plasminogen Activator/administration & dosage , Tumor Suppressor Proteins/immunology
19.
J Neuroinflammation ; 11: 79, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24742325

ABSTRACT

BACKGROUND: Stroke is accompanied by a distinguished inflammatory reaction that is initiated by the infiltration of immunocytes, expression of cytokines, and other inflammatory mediators. As natural killer cells (NK cells) are a type of cytotoxic lymphocyte critical to the innate immune system, we investigated the mechanism of NK cells-induced brain injuries after cerebral ischemia and the chemotactic effect of IP-10 simultaneously. METHODS: NK cells infiltration, interferon-gamma (IFN-γ) and IP-10 expression were detected by immunohistochemistry, immunofluorescence, PCR and flow cytometry in human and C57/BL6 wild type mouse ischemic brain tissues. The ischemia area was detected via 2,3,5-triphenyltetrazolium chloride staining. CXCR3 mean fluorescence intensity of isolated NK cells was measured by flow cytometry. The neuronal injury made by NK cells was examined via apoptosis experiment. The chemotactic of IP-10 was detected by migration and permeability assays. RESULTS: In human ischemic brain tissue, infiltrations of NK cells were observed and reached a peak at 2 to 5 days. In a permanent middle cerebral artery occlusion (pMCAO) model, infiltration of NK cells into the ischemic infarct region reached their highest levels 12 hours after ischemia. IFN-γ-positive NK cells and levels of the chemokine IP-10 were also detected within the ischemic region, from 6 hours up to 4 days after pMCAO was performed, and IFN-γ levels decreased after NK cells depletion in vivo. Co-culture experiments of neural cells with NK cells also showed that neural necrosis was induced via IFN-γ. In parallel experiments with IP-10, the presence of CXCR3 indicates that NK cells were affected by IP-10 via CXCR3, and the effect was dose-dependent. After IP-10 depletion in vivo, NK cells decreased. In migration assays and permeability experiments, disintegration of the blood-brain barrier (BBB) was observed following the addition of NK cells. Moreover, in the presence of IP-10 this injury was aggravated. CONCLUSIONS: All findings support the hypothesis that NK cells participate in cerebral ischemia and promote neural cells necrosis via IFN-γ. Moreover, IP-10 intensifies injury to the BBB by NK cells via CXCR3.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Chemokine CXCL10/metabolism , Killer Cells, Natural/physiology , Animals , Animals, Newborn , Antigens, Ly/metabolism , Apoptosis/physiology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/cytology , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/pathology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Natural Cytotoxicity Triggering Receptor 1/metabolism , Receptors, CXCR3/metabolism
20.
Cell Immunol ; 290(2): 217-25, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25086239

ABSTRACT

Extracellular adenosine is an essential negative regulator of immune reactions that acts by signaling via 4 distinct adenosine receptors. We evaluated adenosine receptor expression in Lewis rats presenting with experimental autoimmune myasthenia gravis (EAMG) to determine whether the expression of adenosine receptors are changed in the development and progression of EAMG. Lymphocyte A1AR and A2AAR mRNA and protein levels from lymphocytes harvested from the lymph nodes, spleen, and peripheral blood mononuclear cells (PBMCs) of EAMG rats were decreased. A modest but not significant increase in A2BAR levels was observed in EAMG lymphocytes harvested from lymph nodes and PBMCs. No changes in A3AR expression were observed in lymphocytes harvested from lymph nodes, spleen, or PBMCs following EAMG induction. Results presented in this report showed that the expression levels and the distribution pattern of adenosine receptors were altered in EAMG lymphocytes.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Receptors, Purinergic P1/biosynthesis , Animals , Autoantibodies/blood , Autoantibodies/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL