Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 63: 191-197, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30099287

ABSTRACT

The heme oxygenase (HO) system is an important regulatory arm of the intrinsic cytoprotective and anti-inflammatory system. HO-2 plays an important role in regulating inflammation following injury. The aim of this study was to evaluate the effect of HO-2 overexpression on inflammatory responses. A skin transplantation model, involving the application of skin grafts from wild-type or HO-2 overexpressing mice to BALB/c mice, was used for investigation. HO-2 overexpression suppressed the production of pro-inflammatory cytokines (IL-1ß, TNF-α, and IL-6) in macrophages compared to that in macrophages obtained from control mice. HO-2 overexpression also significantly prolonged the survival of allografted skin. Our findings suggest that HO-2 attenuates inflammatory responses and effectively prolongs skin graft survival.


Subject(s)
Graft Survival/immunology , Heme Oxygenase (Decyclizing)/immunology , Inflammation/immunology , Macrophages/immunology , Skin Transplantation , Allografts , Animals , Cytokines/genetics , Cytokines/immunology , Female , Heme Oxygenase (Decyclizing)/genetics , Mice, Inbred BALB C , Mice, Transgenic , Skin/metabolism
2.
J Surg Res ; 203(1): 15-21, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27338529

ABSTRACT

BACKGROUND: Hypodermin A (HA) is a serine esterase that degrades complement, a key element of the innate immune system. Immunosuppressive properties of HA have previously been studied in vitro. However, such properties have not been fully demonstrated in vivo. The aim of this study was to evaluate the effect of HA in inhibiting allograft rejection in an HA transgenic mouse model. METHODS: FVB (HA transgenic mice or wild-type mice) to BALB/c mice skin transplantation model were used. Skin grafts were analyzed by histology, immunohistochemistry, and Western blotting. RESULTS: HA overexpression resulted in significantly prolonged skin allograft survival. Histologic changes in the skin allografts paralleled the gross appearance of rejection. ELISA and Western blotting showed that HA significantly reduced the content of complement C3 and C9 in HA skin allografts. The expressions of CD4, B7-2, and MHC class II were all significantly suppressed in HA skin allografts compared with the control group. CONCLUSIONS: These findings suggest that HA effectively prolongs skin allograft survival. The study results provide insight into a promising strategy to improve the survival of grafts in humans.


Subject(s)
Genetic Therapy/methods , Graft Rejection/prevention & control , Graft Survival/immunology , Serine Endopeptidases/immunology , Skin Transplantation , Animals , Biomarkers/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Graft Rejection/enzymology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Transgenic , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Transplantation, Homologous , Treatment Outcome , Up-Regulation
3.
Brain Res ; 1631: 204-13, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26581336

ABSTRACT

The neurotrophic factor insulin-like growth factor (IGF)-1 promotes neurogenesis in the mammalian brain and provides protection against brain injury. However, studies regarding the effects of IGF-1 on cognitive function in aged mice remain limited. We investigated the effects of overexpression of IGF-1 specifically in neural stem cells of the hippocampal dentate gyrus on the recognitive function in 18-month-old transgenic mice. Immunohistocytochemistry and Nissl staining revealed the increased population of BrdU-positive cells as well as the upregulated expression of Nestin and neuronal nuclei (NeuN), respective markers for neural progenitors and neurons, in the hippocampus of the aged IGF-1 transgenic mice versus the wild-type, suggesting that IGF-1 overexpression promotes neurogenesis. In addition, the IGF-1 receptor (IGF-1R), the phosphorylation of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) were enhanced in the transgenic mice than in the wild-type. Transgenic mice also showed superior performance in the Morris water maze and step-down memory tests to their wild-type counterparts. Moreover, the learning and memory abilities of transgenic mice were significantly undermined with the blockage of CaMKII and ERK signaling pathway. Accordingly, our findings indicated that IGF-1 may mitigate the aged-associated cognitive decline via promoting neurogenesis in the hippocampus and activating CaMKII and ERK signaling by binding with IGF-1R.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cognition/physiology , Insulin-Like Growth Factor I/biosynthesis , MAP Kinase Signaling System/drug effects , Animals , Dentate Gyrus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Hippocampus/metabolism , Maze Learning , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/metabolism , Neurogenesis , Neurons/metabolism , Phosphorylation , Receptor, IGF Type 1/metabolism , Signal Transduction , Up-Regulation
4.
Mol Neurobiol ; 51(2): 512-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24777577

ABSTRACT

Growth factors like insulin-like growth factor 1 (IGF-1) is reported to mediate neurogenesis in the subgranular zone (SGZ) and the subventricular zone (SVZ) of the adult mammalian brain, but its regulatory mechanism remains unclear. We generated transgenic mice overexpressing IGF-1 specifically in neural stem cells (NSCs) and assessed the effect of IGF-1 on neurogenesis in adult mice NSCs. Overexpression of IGF-1 could stimulate the expression of phospho-Akt and phospho-ERK1/2 while inducing proliferation and differentiation of NSCs in the SGZ and SVZ. The MEK/ERK inhibitor U0126 could inhibit ERK1/2 phosphorylation, further inhibiting the proliferation of NSCs in the SGZ and SVZ but had no effect on the phosphorylation of Akt. By contrast, The PI3K/Akt inhibitor LY294002 inhibited phosphorylation of Akt and differentiation of NSCs in the SGZ and SVZ, resulting in no change in the proliferation of NSCs and ERK1/2 phosphorylation. These results demonstrate that IGF-1 upregulates the proliferation of NSCs by triggering MEK/ERK pathway signaling in the adult mice SGZ and SVZ. Meanwhile, IGF-1 also induces differentiation of NSCs via the PI3K/Akt pathway in adult mice. However, we found no evidence of crosstalk between the PI3K/Akt and MEK/ERK pathways in adult mice NSCs. Our work provides new experimental evidence of the involvement of the PI3K/Akt and MEK/ERK pathways in the proliferation and differentiation of the NSCs of adult mice.


Subject(s)
Insulin-Like Growth Factor I/biosynthesis , Neural Stem Cells/physiology , Neurogenesis/physiology , Adult Stem Cells/physiology , Animals , Cell Proliferation/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Mol Neurobiol ; 50(3): 971-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24788682

ABSTRACT

Heme oxygenase (HO) represents an intrinsic antiinflammatory system based on its ability to inhibit expression of proinflammatory cytokines. The constitutive isoform heme oxygenase-2 (HO-2) has high expression and activity in cerebral microvascular endothelial cells (CMVEC). This study was undertaken to evaluate the role of HO-2 in regulation of TLR4/MyD88-dependent signaling and to study the effect of HO-2 on the expression and secretion of the proinflammatory cytokines tumor necrosis factor α (TNF-α) and Interleukin-6 (IL6) in CMVEC. HO-2 short hairpin RNA (shRNA) and HO-2 overexpression plasmids were used to observe the effect of HO-2 on proinflammatory cytokines in CMVEC in vitro, and the results showed that the messenger RNA (mRNA) and protein levels of TNF-α and IL6 were increased and decreased, respectively, compared with control groups. LPS-stimulated TNF-α and IL6 mRNA and protein were also reduced in CMVEC treated with an inhibitor of TLR4 signaling, CLI-095, or HO-2 overexpression. CLI-095 and HO-2 overexpression both reduced TLR4 expression in CMVEC, and HO-2 shRNA blocked these effects of CLI-095. CLI-095 and HO-2 overexpression potently suppressed TLR4/MyD88-dependent proinflammatory cytokine expression in CMVEC. These results suggest that HO-2 plays an important role in protecting CMVEC against cytokine-mediated inflammation.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Interleukin-6/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Heme Oxygenase (Decyclizing)/genetics , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Mice , RNA, Small Interfering , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tumor Necrosis Factor-alpha/genetics
6.
Parasitol Int ; 63(2): 392-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24412715

ABSTRACT

Hypodermin A (HA), a serine protease secreted by first-instar larvae of Hypoderma lineatum (Diptera: Oestridae) is associated with inflammatory and the specific immune responses in cattle hosts. In the present study, the cDNA sequence of HA was synthesized, and found to have fifteen amino acids which differed from the sequence available in GenBank. We then examined the association between recombinant HA and guinea-pig complement component 3 (C3) through a co-immunoprecipitation assay. Cos7 cells stably expressing HA were generated, and were found to be more resistant to lysis by guinea-pig C3 than the controls. HA was also able to degrade the C6 and C5b-9 of guinea-pig C3. The presumed DNA binding site of HA with guinea-pig C3 was detected by an electrophoretic mobility shift assay (EMSA). In contrast, after stable transfection, mHA was unable to reduce the amount of C3 or to inhibit its cytotoxicity, while HA could degrade guinea-pig C3 and inhibit the complement pathway. The findings suggest that recombinant HA could serve as an immunosuppressive agent against organ rejection after xenotransplantation.


Subject(s)
Complement System Proteins/metabolism , Serine Endopeptidases/pharmacology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , DNA, Complementary/genetics , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...