Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
J Neuroimmune Pharmacol ; 16(2): 363-375, 2021 06.
Article in English | MEDLINE | ID: mdl-32504312

ABSTRACT

Alzheimer's disease (AD) is an irreversible progressive neurodegenerative disorder recognized by accumulation of amyloid-plaques (APs) and neurofibrillary tangles (NFTs) and eventually loss of memory. Glia maturation factor (GMF), a neuroinflammatory protein first time isolated and cloned in our laboratory plays an important role in the pathogenesis of AD. However, no studies have been reported on whether anti-GMF antibody administration could downregulate neuroinflammation and attenuate amyloid pathology in AD brain. We investigated the potential effect of single dose of (2 mg/kg b.wt/mouse) intravenously (iv) injected with anti-GMF antibodyon cognitive function, neuroprotection, neuroinflammation and Aß load in the brain of 9-month-old 5XFAD mice. Following 4 weeks of anti-GMF antibody delivery in mice, we found reduced expression of GMF, astrocytic glial fibrillary acidic protein (GFAP) and microglial ionizing calcium binding adaptor molecule 1 (Iba1) as well as improvement inneuroinflammatory response via inhibition of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) production and amyloid pathology in the cerebral cortex and hippocampal CA1 region of 5XFAD mice. Correspondingly, blockade of GMF function with anti-GMF antibody improved spatial learning, memory, and long-term recognition memory in 5XFAD mice. The present study demonstrates that the immune checkpoint blockade of GMF function with anti-GMF antibody coordinates anti-inflammatory effects to attenuate neurodegeneration in the cortex and hippocampal CA1 region of 5XFAD mouse brain. Further, our data suggest, that pharmacological immune neutralization of GMF is a promising neuroprotective strategy totherapeutically target neuroinflammation and neurodegeneration in AD. Graphical Abstract 5XFAD mice Polyclonal anti-GMF antibody.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Glia Maturation Factor/antagonists & inhibitors , Plaque, Amyloid/pathology , Animals , Behavior, Animal , Disease Models, Animal , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/pathology
2.
Oncol Rep ; 37(2): 929-936, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28075454

ABSTRACT

Glia maturation factor γ (GMFG) functions to reorganize the actin cytoskeleton and appears to play a causative role in cell migration and adherence. The present study assessed GMFG expression in colorectal cancer cells and tissue specimens and then explored the role of GMFG in colorectal cancer progression in vitro. GMFG protein was highly expressed in colorectal cancer tissues and a metastatic colon cancer cell line. Knockdown of GMFG expression using GMFG siRNA or anti-GMFG antibody decreased the capacity of colon cancer LoVo cell migration and invasion in vitro, while recombinant GMFG treatment induced LoVo cell migration. Furthermore, GMFG knockdown also decreased expression of MMP2 protein and reversed epithelial-mesenchymal transition (EMT) phenotypes in LoVo cells. Co-culture of LoVo cells with human umbilical vein endothelial cells (HUVECs) and exogenous GMFG treatment promoted LoVo cell migration and invasion. The data from the present study indicate that GMFG should be further evaluated as a biomarker for detection of colorectal cancer metastasis and that the targeting of GMFG expression or function could be a novel strategy in the future control of colorectal cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Colorectal Neoplasms/secondary , Gene Expression Regulation, Neoplastic , Glia Maturation Factor/metabolism , Apoptosis , Blotting, Western , Cell Proliferation , Colorectal Neoplasms/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Glia Maturation Factor/antagonists & inhibitors , Glia Maturation Factor/genetics , Humans , Immunoenzyme Techniques , In Vitro Techniques , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
3.
Mol Med Rep ; 10(2): 1122-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24866280

ABSTRACT

ß-elemene, a plant-derived drug extracted from Curcuma wenyujin, has demonstrated marked antiproliferative effects on glioblastoma, while toxicity remains low. However, the underlying molecular mechanisms of the antitumor activity of ß-elemene remain to be elucidated. Previously, it was identified that the glia maturation factor ß (GMFß)/mitogen-activated protein kinase kinase (MAPK) 3/6/p38 pathway participates in the antiproliferative activity of ß-elemene on glioblastoma. In the present study, in order to illustrate the association of GMFß and the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, U87 and U251 cells were treated with ß-elemene at various doses and for different durations, and the expression of phosphorylated ERK1/2 (p-ERK1/2), ERK1/2, B-cell lymphoma 2 (Bcl-2), Bcl2-associated X and survivin was examined by western blot analysis. Following treatment with ß-elemene and the ERK1/2 inhibitor PD98059, U87 cell viability was evaluated using a Cell Counting Kit-8 (CCK-8) assay, and the expression levels of Bcl-2 and survivin were examined by western blot analysis. GMFß was then downregulated by RNA interference in ß-elemene-treated U87 cells, and the effect of this on the expression of ERK1/2 and p-ERK1/2 was determined by western blot analysis. Finally, the chemosensitisation of U87 cells to temozolomide (TMZ) through ß-elemene was examined using the CCK-8 assay. The results demonstrated that ß-elemene inhibited the proliferation of U87 glioblastoma cells through the GMFß­dependent inactivation of the ERK1/2-Bcl-2/survivin pathway. Furthermore, inhibition of ERK1/2 by PD98059 enhanced the antitumor effect of ß-elemene and impaired the expression levels of Bcl-2 and survivin. ß-elemene also increased the sensitivity of U87 glioblastoma cells to the chemotherapeutic TMZ, which was synergistically enhanced by PD98059. In conclusion, these results suggested that GMFß-dependent inactivation of the ERK1/2-Bcl-2/survivin pathway mediated the antiproliferative effect of ß-elemene on glioblastoma. Therefore, ß-elemene is a promising chemosensitizer or adjuvant therapeutic for TMZ against glioblastoma brain tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Cell Proliferation/drug effects , Glia Maturation Factor/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Sesquiterpenes/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Curcuma/chemistry , Curcuma/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/toxicity , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Glia Maturation Factor/antagonists & inhibitors , Glia Maturation Factor/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Survivin , Temozolomide
4.
Exp Cell Res ; 319(5): 707-17, 2013 Mar 10.
Article in English | MEDLINE | ID: mdl-23333559

ABSTRACT

Angiogenesis is a vital process for proper embryonic development, wound healing, malignant tumor growth and metastasis. Two glia maturation factor genes, glia maturation factor-ß (GMFB) and glia maturation factor-γ (GMFG), presenting different expression patterns and distinct biological functions are found in vertebrates. But, the role of GMFB and GMFG in vascular development remains largely unknown. Here, we showed that both GMFB and GMFG are highly conserved in vertebrates. Whole-mount in situ hybridization and quantitative RT-PCR results revealed that GMFB and GMFG were differently expressed during zebrafish embryogenesis. GMFB was highly enriched in brain and GMFG was predominantly expressed in endothelial cells. By gene specific MO, knockdown of GMFG, but not GMFB, severely disrupted angiogenic sprouting of intersegmental vessels (ISVs), but this angiogenic defects were prevented by overexpression of a MO-resistant form of zebrafish GMFG mRNA. In addition, the expressions of angiogenic factors VEGF-A, STAT3, MMP2, MMP9, and MMP13 were significantly decreased in endothelial cells of GMFG morphants. Our findings provide the first in vivo evidence that GMFG is an important regulator for angiogenic sprouting during angiogenesis in zebrafish and suggest that GMFG may act as a novel potential target for anti-angiogenesis therapy in clinical settings.


Subject(s)
Endothelium, Vascular/cytology , Gene Expression Regulation, Developmental , Glia Maturation Factor/metabolism , Neovascularization, Physiologic , Animals , Blotting, Western , Endothelium, Vascular/metabolism , Glia Maturation Factor/antagonists & inhibitors , Glia Maturation Factor/genetics , In Situ Hybridization , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
J Leukoc Biol ; 90(3): 529-38, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21653232

ABSTRACT

Chemotaxis is fundamental to the directional migration of neutrophils toward endogenous and exogenous chemoattractants. Recent studies have demonstrated that ADF/cofilin superfamily members play important roles in reorganizing the actin cytoskeleton by disassembling actin filaments. GMFG, a novel ADF/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in regulating actin reorganization in microendothelial cells, but its function in neutrophils remains unclear. Here, we show that GMFG is an important regulator for cell migration and polarity in neutrophils. Knockdown of endogenous GMFG impaired fMLF- and IL-8 (CXCL8)-induced chemotaxis in dHL-60 cells. GMFG knockdown attenuated the formation of lamellipodia at the leading edge of cells exposed to fMLF or CXCL8, as well as the phosphorylation of p38 and PAK1/2 in response to fMLF or CXCL8. Live cell imaging revealed that GMFG was recruited to the leading edge of cells in response to fMLF, as well as CXCL8. Overexpression of GMFG enhanced phosphorylation of p38 but not of PAK1/2 in dHL-60 cells. In addition, we found that GMFG is associated with WAVE2. Taken together, our findings suggest that GMFG is a novel factor in regulating neutrophil chemotaxis by modulating actin cytoskeleton reorganization.


Subject(s)
Cell Movement , Chemotaxis , Glia Maturation Factor/metabolism , Neutrophils/metabolism , Pseudopodia/metabolism , Actins/metabolism , Adult , Blotting, Western , Cell Adhesion , Cell Polarity , Cell Proliferation , Cells, Cultured , Cytoskeleton/metabolism , Fluorescent Antibody Technique , Glia Maturation Factor/antagonists & inhibitors , Glia Maturation Factor/genetics , Humans , Immunoprecipitation , Interleukin-8/pharmacology , Neutrophils/immunology , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL