Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 11(9)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34575110

ABSTRACT

Despite the fact that accumulation of microglia, the resident macrophages of the central nervous system (CNS) are the main feature of glioblastoma, the role of microglia in the progression of glioma is still arguable. Based on the correlation of inflammation with tumor progression, in this study, we attempt to determine if peripheral inflammation aggravates glioma expansion and the activation of microglia associated with the tumor. Experimental animals were administered intraperitoneally by inflammagen lipopolysaccharide (LPS) for 7 days (LPS priming) before intracerebral implantation of glioma cells. Moreover, a reduced level of tumor necrosis factor receptor type 2 (TNFR2) that is restricted to immune cells, neurons, and microglia has been found in patients with glioblastoma through the clinic analysis of monocyte receptor expression. Thus, in addition to wildtype (WT) mice, heterogeneous TNFR2 gene deficiency (TNFR2+/-) mice and homogeneous TNFR2 gene knockout (TNFR2-/-) mice were used in this study. The results show that peripheral challenge by LPS, Iba1+- or CD11b+-microglia increase in numbers in the cortex and hippocampus of TNFR2-/- mice, when compared to WT or TNFR2+/- mice. We further conducted the intracerebral implantation of rodent glioma cells into the animals and found that the volumes of tumors formed by rat C6 glioma cells or mouse GL261 glioma cells were significantly larger in the cortex of TNFR2-/- mice when compared to that measured in LPS-primed WT or LPS-primed TNFR2+/- mice. Ki67+-cells were exclusively clustered in the tumor of LPS-primed TNFR2-/- mice. Microglia were also extensively accumulated in the tumor formed in LPS-primed TNFR2-/- mice. Accordingly, our findings demonstrate that aggravation of microglia activation by peripheral inflammatory challenge and a loss of TNFR2 function might lead to the promotion of glioma growth.

2.
Front Mol Neurosci ; 11: 4, 2018.
Article in English | MEDLINE | ID: mdl-29416501

ABSTRACT

B-cell CLL/lymphoma 11B (Bcl11b) - a C2H2 zinc finger transcriptional factor - is known to regulate neuronal differentiation and function in the development of the central nervous system (CNS). Although its expression is reduced during oligodendrocyte (OLG) differentiation, its biological role in OLGs remains unknown. In this study, we found that the downregulation of Bcl11b gene expression in glial progenitor cells (GPCs) by lentivirus-mediated gene knockdown (KD) causes a reduction in cell proliferation with inhibited expression of stemness-related genes, while increasing the expression of cell cyclin regulator p21. In contrast, OLG specific transcription factors (Olig1) and OLG cell markers, including myelin proteolipid protein (PLP) and myelin oligodendrocyte glycoprotein (MOG), were upregulated in Bcl11b-KD GPCs. Chromatin immunoprecipitation (ChIP) analysis indicated that Bcl11b bound to the promoters of Olig1 and PLP, suggesting that Bcl11b could act as a repressor for Olig1 and PLP, similar to its action on p21. An increase in the number of GC+- or PLP+- OLGs derived from Bcl11b-KD GPCs or OLG precursor cells was also observed. Moreover, myelin basic protein (MBP) expression in OLGs derived from Bcl11b-KD GPCs was enhanced in hippocampal neuron co-cultures and in cerebellar brain-slice cultures. The in vivo study using a lysolecithin-induced demyelinating animal model also indicated that larger amounts of MBP+-OLGs and PLP+-OLGs derived from implanted Bcl11b-KD GPCs were present at the lesioned site of the white matter than in the scramble group. Taken together, our results provide insight into the functional role of Bcl11b in the negative regulation of GPC differentiation through the repression of OLG differentiation-associated genes.

3.
Mol Neurobiol ; 55(6): 4624-4636, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28707070

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor subtype. Despite that metastasis of GBM beyond the central nervous system (CNS) is rare, its malignancy is attributed to the highly infiltration trait, leading to the difficulty of complete surgical excision. Matrix gla protein (MGP) is a vitamin K-dependent small secretory protein, and functions as a calcification inhibitor. The involvement of MGP function in glioma cell dynamics remains to be clarified. The study showed that a low proliferative rat C6 glioma cell line named as C6-2 exhibited faster migratory and invasive capability compared to that observed in a high tumorigenic rat C6 glioma cell line (called as C6-1). Interestingly, C6-2 cells expressed higher levels of MGP molecules than C6-1 cells did. Lentivirus-mediated short hairpin RNA (shRNA) against MGP gene expression (MGP-KD) in C6-2 cells or lentivirus-mediated overexpression of MGP transcripts in C6-1 cells resulted in the morphological alteration of the two cell lines. Moreover, MGP-KD caused a decline in cell migration and invasion ability of C6-2 cells. In contrast, increased expression of MGP in C6-1 cells promoted their cell migration and invasion. The observations were further verified by the results from the implantation of C6-1 and C6-2 cells into ex vivo brain slice and in vivo rat brain. Thus, our results demonstrate that the manipulation of MGP expression in C6 glioma cells can mediate glioma cell migratory activity. Moreover, our findings indicate the possibility that high proliferative glioma cells expressing a high level of MGP may exist and contribute to tumor infiltration and recurrence.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Calcium-Binding Proteins/metabolism , Cell Movement , Extracellular Matrix Proteins/metabolism , Glioma/metabolism , Glioma/pathology , Animals , Brain Neoplasms/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Neoplasm Invasiveness , Rats, Sprague-Dawley , Up-Regulation , Matrix Gla Protein
4.
J Neurosci Res ; 94(12): 1460-1471, 2016 12.
Article in English | MEDLINE | ID: mdl-27629530

ABSTRACT

CD200, a type I transmembrane glycoprotein, can interact with its receptor CD200R, which plays an inhibitory role in the activation of microglia-the resident macrophages of the central nervous system. In this study, the rat C6 glioma cell line (C6-1) that was previously characterized with high in vivo tumorigenicity was found to generate CD200 mRNA abundantly. However, CD200 expression was barely detected in another C6 glioma cell clone (C6-2) that was previously found to display low tumorigenic behavior. The results from CD200 immunohistochemistry on human glioma tissue array also showed that tumor cells in Grade I-II astrocytoma expressed a lower level of CD200 immunoreactivity than those detected in Grade III-IV glioblastoma multiforme. C6-1 transfectants with stable downregulation of CD200 gene expression using lentivirus knockdown approach were generated (C6-KD). Microglia and iNOS+ cells were increased when microglia were co-cultured with C6-KD cells. The colony formation of C6-KD was also augmented when those cells were co-cultured with microglia. Yet, increased colony formation of C6-KD transfectants in the co-culture with microglia was effectively suppressed by interleukin (IL)-4 and IL-10. The in vivo results indicated that the tumor formation of C6-1 cells in rat brain was promoted after CD200 gene knockdown. Moreover, CD11b+ activated microglia and iNOS+ microglia were highly accumulated in the tumor site formed by C6-KD. In conclusion, our findings demonstrate that the downregulation of CD200 expression in CD200-rich glioma cells could foster the formation of an activated microglia-associated tumor microenvironment, leading to glioma progression. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antigens, CD/biosynthesis , Brain Neoplasms/metabolism , Glioma/metabolism , Macrophage Activation , Microglia , Animals , Antigens, CD/genetics , Astrocytoma/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioma/pathology , Humans , Immunohistochemistry , Interleukin-10/pharmacology , Interleukin-4/pharmacology , Male , Nitric Oxide Synthase Type II/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
5.
Mol Neurobiol ; 53(6): 3528-3539, 2016 08.
Article in English | MEDLINE | ID: mdl-26096706

ABSTRACT

B cell CLL/lymphoma 11B (Bcl11b), a C2H2 zinc finger transcription factor, not only serves as a critical regulator in development but also plays the controversial role in T cell acute lymphoblastic leukemia (T-ALL). We previously found that the enriched expression of Bcl11b was detected in high tumorigenic C6 glioma cells. However, the role of Bcl11b in glioma malignancy and its mechanisms remains to be uncovered. In this study, using the lentivirus-mediated knockdown (KD) approach, we found that Bcl11b KD in tumorigenic C6 cells reduced the cell proliferation, colony formation, and migratory ability. The results were further verified using two human malignant glioma cell lines, U87 and U251 cells. A cyclin-dependent kinase inhibitor p21, a known Bcl11b target, was significantly upregulated in tumorigenic C6, U87, and U251 cells after Bcl11b KD. Cellular senescence was observed by examination of the ß-galactosidase activity in U87 and U251 cells with Bcl11b KD. Reduced expression of stemness gene Sox-2 and its downstream effector Bmi-1 was also observed in U87 and U251 cells with Bcl11b KD. These results suggest that the ablation of Bcl11b gene expression induced glioma cell senescence. Propidium iodide (PI) staining combined with flow cytometry analysis also showed that Bcl11b KD led to the cell cycle arrest of U87 and U251 cells at the G0/G1 or at the S phase, indicating that Bcl11b is required for glioma cell cycle progression. Together, this is the first study to show that the inhibition of Bcl11b suppresses glioma cell growth by regulating the expression of the cell cycle regulator p21 and stemness-associated genes (Sox-2/Bmi-1).


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Brain Neoplasms/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/genetics , Gene Knockdown Techniques , Humans , Polycomb Repressive Complex 1/metabolism , Rats , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation/genetics
6.
J Neurooncol ; 124(3): 403-12, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26239968

ABSTRACT

Colchicine, an anti-microtubule and antimitotic drug, is a common therapeutically agent for gout, which is thought to have potential anti-tumor effects. Owing to concerns of colchicines poisoning, the development of derivatives with low dose efficacy and less side effects is of obvious interest. In this study, we characterized the inhibitory effects of a colchicine derivative named AD1 on the cell proliferation of human malignant glioblastoma (MG) cell lines, U87MG and U373MG. We found that 50 % of U87MG and U373MG cells were reduced in the cultures after exposure to AD1 for 24 h at 10 and 50 nM, respectively. Moreover, α-tubulin immunostaining indicated that AD1 induced the disruption of the microtubule polymerization in glioma cells with apoptotic features including membrane budding/blebbing or fragmented nuclei. Increased levels of reactive oxygen species (ROS) were also detected in AD1-treated U87MG and U373MG cells compared to that observed in the control culture. Moreover, examination of microtubule-associated protein 1A/1B-light chain 3 (LC3I)/LC3II conversion and acridine orange staining for autophagic vesicles, combined with flow cytometry, showed that treatment with AD1 induced the autophagic pathway in U87MG and U373MG cells. Furthermore, we found that the intermittent intravenous administration of AD1 suppressed glioma growth in rat brain receiving intracerebral injection with rat C6 glioma cells. Taken together, our findings reveal that treatment with AD1 at nanomolar scales can reduce glioma cell viability effectively, with the occurrence of a rise in ROS and cellular autophagy. In conjunction with the observations from in vivo study, the colchicine derivative AD1 has chemotherapeutic potential to suppress glioma progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Colchicine/therapeutic use , Glioblastoma/drug therapy , Animals , Autophagy/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Colchicine/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glioblastoma/pathology , Humans , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Time Factors , Tubulin/metabolism , Xenograft Model Antitumor Assays
7.
Oncotarget ; 5(21): 10901-15, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25337721

ABSTRACT

We previously found that BRCA1-BRCA2-containing complex subunit 3 (BRCC3) was highly expressed in tumorigenic rat glioma cells. However, the functional role of BRCC3 in human glioma cells remains to be characterized. This study indicated that the upregulation of BRCC3 expression was induced in two human malignant glioblastoma U251 and A172 cell lines following exposure to the alkylating agent, temozolomide (TMZ). Homologous recombination (HR)-dependent DNA repair-associated genes (i.e. BRCA1, BRCA2, RAD51 and FANCD2) were also increased in U251 and A172 cells after treatment with TMZ. BRCC3 gene knockdown through lentivirus-mediated gene knockdown approach not only significantly reduced the clonogenic and migratory abilities of U251 and A172 cells, but also enhanced their sensitization to TMZ. The increase in phosphorylated H2AX foci (γH2AX) formation, an indicator of DNA damage, persisted in TMZ-treated glioma cells with stable knockdown BRCC3 expression, suggesting that BRCC3 gene deficiency is associated with DNA repair impairment. In summary, we demonstrate that by inducing DNA repair, BRCC3 renders glioma cells resistant to TMZ. The findings point to BRCC3 as a potential target for treatment of alkylating drug-resistant glioma.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Membrane Proteins/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Deubiquitinating Enzymes , Down-Regulation , Fluorescent Antibody Technique , Glioma/metabolism , Glioma/pathology , Humans , Immunoenzyme Techniques , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Neoplasm Grading , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Temozolomide , Tumor Cells, Cultured
8.
Neuro Oncol ; 16(4): 552-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24327583

ABSTRACT

BACKGROUND: Glioma development is a multistep process associated with progressive genetic alterations but also regulated by cellular and noncellular components in a tumor-associated niche. METHODS: Using 2 rat C6 glioma cell clones with different tumorigenesis, named C6-1 and C6-2, this study characterized genes associated with enhanced tumorigenic features of glioma cells by comparative cDNA microarray analysis combined with Q-PCR. Neurospehere formation and clonogenicity were examined to determine the growth of tumorigenic C6 glioma cells. The lentivirus-mediated gene knockdown approach was conducted to determine the role of interleukin-33 (IL-33) in glioma cell proliferation and migration. Transwell cell invasion assay was used to examine microglia migration induced by tumorigenic C6 cells. RESULTS: The functional analysis of gene ontology (GO) biological processes shows that the upregulated genes found in tumorigenic C6 (C6-1) cells are closely related to cell proliferation. Tumorigenic C6 cells expressed cytokines and chemokines abundantly. Among these genes, IL-33 was profoundly induced in tumorigenic C6 cells with the expression of IL-33 receptor ST2. Furthermore, the growth rate and colony formation of tumorigenic C6 cells were attenuated by the inhibition of IL-33 and ST2 gene expression. Moreover, IL-33 was involved in tumorigenic glioma cell migration and regulation of the expression of several glioma-associated growth factors and chemokines in tumorigenic C6 cells. CONCLUSION: Accordingly, we concluded that glioma cells with abundant production of IL-33 grow rapidly; moreover, the interactions of multiple cytokines/chemokines induced by glioma cells may develop a microenvironment that facilitates microglia/macrophage infiltration and fosters glioma growth in the brain.


Subject(s)
Cell Movement , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Interleukins/metabolism , Animals , Animals, Newborn , Apoptosis , Blotting, Western , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Glioma/genetics , Immunoenzyme Techniques , Interleukin-33 , Interleukins/antagonists & inhibitors , Interleukins/genetics , Lentivirus/genetics , Mice , Microglia/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Glia ; 61(9): 1402-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23832679

ABSTRACT

CD133 (Prominin-1/AC133) is generally treated as a cell surface marker found on multipotent stem cells and tumor stem-like cells, and its biological function remains debated. Genetically modified rat glioma cell lines were generated by lentiviral gene delivery of human CD133 into rat C6 glioma cells (hCD133(+) -C6) or by infection of C6 cells with control lentivirus (mock-C6). Stable hCD133 expression promoted the self-renewal ability of C6-formed spheres with an increase in the expression of the stemness markers, Bmi-1 and SOX2. Akt phosphorylation, Notch-1 activation, and Notch-1 target gene expression (Hes-1, Hey1 and Hey2) were increased in hCD133(+) -C6 when compared to mock-C6. The inhibition of Akt phosphorylation, Notch-1 activation, and Hes-1 in hCD133(+) -C6 cells effectively suppressed their clonogenic ability, indicating that these factors are involved in expanding the growth of hCD133(+) -C6. An elevated expression of GTPase-activating protein 27 (Arhgap27) was detected in hCD133(+) -C6. A decline in the invasion of hCD133(+) -C6 by knockdown of Arhgap27 expression indicated the critical role of Arhgap27 in promoting cell migration of hCD133(+) -C6. In vivo study further showed that hCD133(+) -C6 formed aggressive tumors in vivo compared to mock-C6. Exposure of hCD133(+) -C6 to arsenic trioxide not only reduced Akt phosphorylation, Notch-1 activation and Hes-1 expression in vitro, but also inhibited their tumorigenicity in vivo. The results show that C6 glioma cells with stable hCD133 expression enhanced their stemness properties with increased Notch-1/Hes-1 signaling, Akt activation, and Arhgap27 action, which contribute to increased cell proliferation and migration of hCD133(+) -C6 in vitro, as well as progressive tumor formation in vivo.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Glioma/metabolism , Glycoproteins/metabolism , Peptides/metabolism , AC133 Antigen , Animals , Antigens, CD/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Arsenic Trioxide , Arsenicals/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Formazans , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glioma/drug therapy , Glioma/pathology , Glycoproteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Lentivirus/genetics , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Oxides/pharmacology , Peptides/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Tetrazolium Salts , Time Factors , Transcription Factor HES-1 , Transfection
10.
J Cell Biochem ; 113(9): 3019-28, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22566124

ABSTRACT

Astrocytes, the major glial population in the central nervous system (CNS), play an important role in neuronal homeostasis, neurogenesis, and synaptogenesis. The cells have a stellate shape with elaborated processes in the developing CNS. Cultured astrocytes become stellate when the cells undergo differentiation in response to stimuli. Nevertheless, the molecular mechanism for astrocytic stellation is poorly understood. Here, we showed that the addition of serum induced a flat polygonal shape in cultured astrocytes with a reduced level of Mps one binder 2 (Mob2) that is involved in neurite growth by forming stable complex with a nuclear Ser/Thr kinase Dbf2-related protein kinase 1 (NDR1). Furthermore, exposure to a membrane permeable cAMP analogue, dbcAMP, not only induced astrocytic stellation, but also caused an increase in Mob2 expression. Similarly, the upregulation of Mob2 mRNA expression was induced by exposure of astrocytes to pituitary adenylyl cyclase-activating polypeptide (PACAP). Pretreatment with a cAMP/protein kinase A (PKA) inhibitor, KT-5720, significantly blocked the effect of dbcAMP and PACAP on induced upregulation of Mob2 mRNA expression in astrocytes. In addition, the process withdrawal of dbcAMP-treated astrocytes was caused by the inhibition of Mob2 expression using lentivirus-mediated Mob2 shRNA delivery system. Based on our findings, we suggest that Mob2 is involved in PKA signaling-mediated astrocytic stellation.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Cyclic AMP/metabolism , Nerve Tissue Proteins/metabolism , Animals , Blotting, Western , Bucladesine/pharmacology , Carbazoles/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Fluorescent Antibody Technique , Nerve Tissue Proteins/genetics , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
11.
J Neurosci Res ; 89(2): 199-211, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21162127

ABSTRACT

Chemokines can be produced by gliomas, which mediate the infiltration of microglia, a characteristic feature of glioma-associated neuropathogenesis. ATP that is released at a high level from glioma has been reported to play a regulatory role in chemokine production in cultured glioma cells. The objective of this study was to define the potential role of extracellular ATP in the regulation of macrophage inflammatory protein-1α (MIP-1α) and monocyte chemoattractant protein-1(MCP-1) expression in glioma-associated microglia/macrophages. The results showed that Iba1(+) and ED1(+) microglia existed in the tumor at 3 and 7 day after injection of C6 glioma cells into the rat cerebral cortex (dpi). ED1(+) microglia/macrophages or Iba1(+) microglia in the glioma were also colocalized to MIP-1α- and MCP-1-expressing cells. In vitro study indicated that treatment with ATP and BzATP (an agonist for ATP ionotropic receptor P2X7R) caused an increase in the intracellular levels of microglial MIP-1α and MCP-1. By using an extracellular Ca(2+) chelator (EGTA) and P2X7R antagonists, oxidized ATP (oxATP) and brilliant blue G (BBG), we demonstrated that BzATP-induced production of MIP-1α and MCP-1 levels was due to P2X7R activation and Ca(2+) -dependent regulation. Coadministration of C6 glioma cells and oxATP into the rat cerebral cortex resulted in a reduction of MIP-1α- and MCP-1-expressing microglia/macrophages. We suggest, based on the results from in vivo and in vitro studies, that a massive amount of ATP molecules released in the glioma tumor site may act as the regulator with P2X7R signaling that increases MIP-1α and MCP-1 expression in tumor-infiltrating microglia/macrophages.


Subject(s)
Adenosine Triphosphate/metabolism , Chemokine CCL2/biosynthesis , Chemokine CCL3/biosynthesis , Glioma/metabolism , Microglia/metabolism , Receptors, Purinergic P2X/metabolism , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation , Glioma/immunology , Immunohistochemistry , Rats , Rats, Sprague-Dawley
12.
PLoS One ; 5(12): e15299, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21187959

ABSTRACT

The aim of this study is to understand if human mesenchymal stem cells (hMSCs) and neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) have synergistic protective effect that promotes functional recovery in rats with severe spinal cord injury (SCI). To evaluate the effect of delayed combinatorial therapy of PACAP and hMSCs on spinal cord tissue repair, we used the immortalized hMSCs that retain their potential of neuronal differentiation under the stimulation of neurogenic factors and possess the properties for the production of several growth factors beneficial for neural cell survival. The results indicated that delayed treatment with PACAP and hMSCs at day 7 post SCI increased the remaining neuronal fibers in the injured spinal cord, leading to better locomotor functional recovery in SCI rats when compared to treatment only with PACAP or hMSCs. Western blotting also showed that the levels of antioxidant enzymes, Mn-superoxide dismutase (MnSOD) and peroxiredoxin-1/6 (Prx-1 and Prx-6), were increased at the lesion center 1 week after the delayed treatment with the combinatorial therapy when compared to that observed in the vehicle-treated control. Furthermore, in vitro studies showed that co-culture with hMSCs in the presence of PACAP not only increased a subpopulation of microglia expressing galectin-3, but also enhanced the ability of astrocytes to uptake extracellular glutamate. In summary, our in vivo and in vitro studies reveal that delayed transplantation of hMSCs combined with PACAP provides trophic molecules to promote neuronal cell survival, which also foster beneficial microenvironment for endogenous glia to increase their neuroprotective effect on the repair of injured spinal cord tissue.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cells/cytology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Spinal Cord/pathology , Animals , Astrocytes/metabolism , Cell Survival , Glutamic Acid/metabolism , Humans , Microglia/metabolism , Models, Biological , Rats , Superoxide Dismutase/metabolism
13.
J Neurochem ; 111(5): 1225-37, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19860838

ABSTRACT

Microglia, the CNS resident macrophages responsible for the clearance of degenerating cellular fragments, are essential to tissue remodeling and repair after CNS injury. ATP can be released in large amounts after CNS injury and may mediate microglial activity through the ionotropic P2X and the metabotropic P2Y receptors. This study indicates that exposure to a high concentration of ATP for 30 min rapidly induces changes of the microglial cytoskeleton, and significantly attenuates microglial phagocytosis. A pharmacological approach showed that ATP-induced inhibition of microglial phagocytotic activity was due to P2X(7)R activation, rather than that of P2YR. Activation of P2X(7)R by its agonist, 2'-3'-O-(4-benzoyl)benzoyl-ATP (BzATP), produced a Ca(2+)-independent reduction in microglial phagocytotic activity. In addition, the knockdown of P2X(7)R expression by lentiviral-mediated shRNA interference or the blockade of P2X(7)R activation by the specific antagonists, oxidized ATP (oxATP) and brilliant blue G, has efficiently restored the phagocytotic activity of ATP and BzATP-treated microglia. Our results reveal that P2X(7)R activation may induce the formation of a Ca(2+)-independent signaling complex, which results in the reduction of microglial phagocytosis. This suggests that exposure to ATP for a short-term period may cause insufficient clearance of tissue debris by microglia through P2X(7)R activation after CNS injury, and that blockade of this receptor may preserve the phagocytosis of microglia and facilitate CNS tissue repair.


Subject(s)
Adenosine Triphosphate/pharmacology , Microglia/physiology , Phagocytosis/drug effects , Receptors, Purinergic P2/metabolism , Actins/metabolism , Adenosine Triphosphate/analogs & derivatives , Animals , Animals, Newborn , Benzoxazoles/metabolism , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cyclophilin A/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry/methods , Microglia/drug effects , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Quinolinium Compounds/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X2 , Time Factors , Transduction, Genetic/methods
14.
J Cell Biochem ; 107(5): 933-43, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19459161

ABSTRACT

Astrocytes, the most abundant glial cell population in the central nervous system (CNS), play physiological roles in neuronal activities. Oxidative insult induced by the injury to the CNS causes neural cell death through extrinsic and intrinsic pathways. This study reports that reactive oxygen species (ROS) generated by exposure to the strong oxidizing agent, hexavalent chromium (Cr(VI)) as a chemical-induced oxidative stress model, caused astrocytes to undergo an apoptosis-like cell death through a caspase-3-independent mechanism. Although activating protein-1 (AP-1) and NF-kappaB were activated in Cr(VI)-primed astrocytes, the inhibition of their activity failed to increase astrocytic cell survival. The results further indicated that the reduction in mitochondrial membrane potential (MMP) was accompanied by an increase in the levels of ROS in Cr(VI)-primed astrocytes. Moreover, pretreatment of astrocytes with N-acetylcysteine (NAC), the potent ROS scavenger, attenuated ROS production and MMP loss in Cr(VI)-primed astrocytes, and significantly increased the survival of astrocytes, implying that the elevated ROS disrupted the mitochondrial function to result in the reduction of astrocytic cell viability. In addition, the nuclear expression of apoptosis-inducing factor (AIF) and endonuclease G (EndoG) was observed in Cr(VI)-primed astrocytes. Taken together, evidence shows that astrocytic cell death occurs by ROS-induced oxidative insult through a caspase-3-independent apoptotic mechanism involving the loss of MMP and an increase in the nuclear levels of mitochondrial pro-apoptosis proteins (AIF/EndoG). This mitochondria-mediated but caspase-3-independent apoptotic pathway may be involved in oxidative stress-induced astrocytic cell death in the injured CNS.


Subject(s)
Astrocytes/cytology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Apoptosis Inducing Factor/metabolism , Astrocytes/drug effects , Astrocytes/enzymology , Caspase 3 , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Chromium/toxicity , Cytoprotection/drug effects , DNA/metabolism , Endodeoxyribonucleases/metabolism , Enzyme Activation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , NF-kappa B/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...