Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(12): 1504-16, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26165844

ABSTRACT

Reactive astrocytes are integral to the glioma microenvironment. Connexin43 (Cx43) is a major gap junction protein in astrocytes and its expression is enhanced significantly in glioma-associated astrocytes, especially at the peri-tumoral region. Although downregulation of Cx43-mediated intercellular communication is associated with increased malignancy in tumor cells, the role of Cx43 in stromal cells in glioma progression is not defined. Using a mouse model consisting of syngeneic intracranial implantation of GL261 glioma cells into Nestin-Cre:Cx43(fl/fl) mice where Cx43 was eliminated in astrocytes, we demonstrate a role of astrocytic Cx43 in the dissemination of glioma cells from the tumor core. To determine whether heterocellular communication between astrocytes and glioma cells is essential for reduced invasion in the absence of astrocytic Cx43, we abolished channel formation between glioma cells and astrocytes by either knocking down Cx43 in glioma cells with short hairpin RNA (shRNA) or overexpressing a dominant-negative channel-defective Cx43-T154A mutant in these cells. Although Cx43shRNA in glioma cells reduced invasion, expression of Cx43-T154A had no effect on glioma invasion, suggesting tumoral Cx43 may influence motility independently from its channel function. Alteration in astrocytic Cx43 function, such as by replacing the wild-type allele with a C-terminal truncated Cx43 mutant exhibiting reduced intercellular coupling, is sufficient to reduce glioma spreading into the brain parenchyma. Our results reveal a novel role of astrocytic Cx43 in the formation of an invasive niche and raise the possibility to control glioma progression by manipulating the microenvironment.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/pathology , Connexin 43/physiology , Glioma/pathology , Neoplasm Invasiveness , Animals , Cell Adhesion , Female , Male , Mice , Mice, Knockout
2.
Neuroscience ; 323: 207-21, 2016 May 26.
Article in English | MEDLINE | ID: mdl-25913636

ABSTRACT

The World Health Organization has predicted that by 2040 neurodegenerative diseases will overtake cancer to become the world's second leading cause of death after cardiovascular disease. This has sparked the development of several European and American brain research initiatives focusing on elucidating the underlying cellular and molecular mechanisms of neurodegenerative diseases. Connexin (Cx) and pannexin (Panx) membrane channel proteins are conduits through which neuronal, glial, and vascular tissues interact. In the brain, this interaction is highly critical for homeostasis and brain repair after injury. Understanding the molecular mechanisms by which these membrane channels function, in health and disease, might be particularly influential in establishing conceptual frameworks to develop new therapeutics against Cx and Panx channels. This review focuses on current insights and emerging concepts, particularly the impact of connexin43 and pannexin1, under neuroprotective and neurodegenerative conditions within the context of astrocytes.


Subject(s)
Astrocytes/metabolism , Connexin 43/metabolism , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neuroprotection/physiology , Animals , Connexin 43/genetics , Humans , Nerve Tissue Proteins/genetics
3.
Cell Death Differ ; 19(6): 947-57, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22117194

ABSTRACT

Decades of research have indicated that gap junction channels contribute to the propagation of apoptosis between neighboring cells. Inositol 1,4,5-trisphosphate (IP3) has been proposed as the responsible molecule conveying the apoptotic message, although conclusive results are still missing. We investigated the role of IP3 in a model of gap junction-mediated spreading of cytochrome C-induced apoptosis. We used targeted loading of high-molecular-weight agents interfering with the IP3 signaling cascade in the apoptosis trigger zone and cell death communication zone of C6-glioma cells heterologously expressing connexin (Cx)43 or Cx26. Blocking IP3 receptors or stimulating IP3 degradation both diminished the propagation of apoptosis. Apoptosis spread was also reduced in cells expressing mutant Cx26, which forms gap junctions with an impaired IP3 permeability. However, IP3 by itself was not able to induce cell death, but only potentiated cell death propagation when the apoptosis trigger was applied. We conclude that IP3 is a key necessary messenger for communicating apoptotic cell death via gap junctions, but needs to team up with other factors to become a fully pro-apoptotic messenger.


Subject(s)
Apoptosis , Gap Junctions/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Cell Communication , Cell Membrane Permeability , Connexin 26 , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Cytochromes c/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Rats , Signal Transduction
4.
Oncogene ; 30(45): 4601-8, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21602884

ABSTRACT

Ionizing radiation is a genotoxic agent and human carcinogen. Recent work has questioned long-held dogmas by showing that cancer-associated genetic alterations occur in cells and tissues not directly exposed to radiation, questioning the robustness of the current system of radiation risk assessment. In vitro, diverse mechanisms involving secreted soluble factors, gap junction intercellular communication (GJIC) and oxidative metabolism are proposed to mediate these indirect effects. In vivo, the mechanisms behind long-range 'bystander' responses remain largely unknown. Here, we investigate the role of GJIC in propagating radiation stress signals in vivo, and in mediating radiation-associated bystander tumorigenesis in mouse central nervous system using a mouse model in which intercellular communication is downregulated by targeted deletion of the connexin43 (Cx43) gene. We show that GJIC is critical for transmission of oncogenic radiation damage to the non-targeted cerebellum, and that a mechanism involving adenosine triphosphate release and upregulation of Cx43, the major GJIC constituent, regulates transduction of oncogenic damage to unirradiated tissues in vivo. Our data provide a novel hypothesis for transduction of distant bystander effects and suggest that the highly branched nervous system, similar to the vascular network, has an important role.


Subject(s)
Adenosine Triphosphate/metabolism , Bystander Effect/radiation effects , Cell Transformation, Neoplastic/genetics , Cerebellar Neoplasms/genetics , Connexin 43/metabolism , DNA Damage/genetics , Neoplasms, Radiation-Induced/genetics , Animals , Cerebellum/metabolism , Cerebellum/radiation effects , Connexin 43/genetics , Gap Junctions/metabolism , Gap Junctions/radiation effects , Mice , Radiation Dosage , Sequence Deletion/radiation effects , Signal Transduction/radiation effects
5.
Oncogene ; 29(42): 5712-23, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20676131

ABSTRACT

One of the characteristics of gliomas is a decrease in the expression of connexin43, a protein that forms gap junctions. Restoring connexin43 expression in glioma cells reduces their exacerbated rate of cell growth, although it is not yet known how connexin43 modifies the expression of genes involved in cell proliferation. Here, we show that restoring connexin43 to C6 glioma cells impedes their progression from G0/G1 to the S phase of the cell cycle by reducing retinoblastoma phosphorylation and cyclin E expression through the upregulation of p21 and p27. Interestingly, connexin43 diminishes the oncogenic activity of c-Src exhibited by glioma cells. By studying a Tyr247 and Tyr265 mutant connexin43, we show that these residues are required for connexin43 to inhibit c-Src activity and cell proliferation. In conclusion, by acting as a substrate of c-Src, connexin43 reduces its oncogenic activity and decreases the rate of glioma cell proliferation, potentially an early step in the antiproliferative effects of connexin43. Although c-Src is known to phosphorylate connexin43, this study provides the first evidence that connexin43 can also inhibit c-Src activity.


Subject(s)
Connexin 43/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, src/genetics , Glioma/genetics , Animals , Blotting, Western , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Cell Separation , Flow Cytometry , Gene Expression , Glioma/metabolism , Mutation , RNA, Small Interfering , Rats , Transfection
6.
Oncogene ; 28(49): 4402-8, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19749789

ABSTRACT

Connexins (Cxs), the gap junction proteins, have been found to be downregulated in many types of cancers including gliomas. By restoring gap junctional communication in cancer cell models, the neoplastic phenotype can be reversed, suggesting Cxs are tumor suppressors. Pannexin2 (Panx2) is a member of the novel gap junction protein family, Panxs, and it has been proposed as a brain-specific protein. Recently, gene array analysis showed an overall reduction of Panx2 in gliomas, and a direct correlation was observed between Panx2 expression and post-diagnosis survival in patients. In this study, we explored the potential inverse correlation between Panx2 and glioma oncogenicity. A decrease or absence of Panx2 expression in a panel of human glioma cell lines was found, whereas an appreciable amount of Panx2 was detected in both human brain and astrocytes. Stable Panx2 expression revealed a flattened morphology and increased cell-cell contacts in rat C6 glioma cells similar to Panx1. However, in contrast to Panx1 and Panx3, Panx2 was predominately detected in the cytoplasm in vesicle-like patterns but not at the plasma membrane. Coexpression of Panx2 and Panx1 did not show colocalization of both Panxs. Strikingly, restoration of Panx2 expression significantly reduced in vitro oncogenicity parameters, including monolayer saturation density and anchorage-independent growth, as well as in vivo tumor growth. This study suggests a role of aberrant Panx2 expression during gliomagenesis, and that Panx2 independently functions as a negative growth regulator without Panx1.


Subject(s)
Cell Proliferation , Connexins/physiology , Glioma/pathology , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Connexins/genetics , Connexins/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Glioma/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Mice , Mice, Nude , Mice, SCID , Mice, Transgenic , Rats , Tissue Distribution , Transplantation, Heterologous
7.
Cell Death Differ ; 16(1): 151-63, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18820645

ABSTRACT

Gap junctions (GJs) have been demonstrated to communicate cell death signals from apoptotic to healthy cells, thereby spatially extending apoptosis. Before being incorporated into GJs, hemichannels (hemi-GJs) are normally closed but recent evidence suggests that they can be opened by various messengers and conditions, thereby forming a pore through which molecules can enter or leave the cell potentially leading to cell death. The aim of this study was to determine the contribution of GJs and hemichannels in the communication of apoptosis toward surrounding cells. We induced apoptosis in C6 glioma cells stably transfected with connexin (Cx)43, with cytochrome C (cytC) using in situ electroporation and found that healthy surrounding cells underwent apoptotic transformation. Work with various cell death markers, wild-type (WT) and Cx43-expressing cells, inhibitors of GJs and/or hemichannels, and Cx43 gene silencing showed that GJs contribute to the spread of apoptosis in a zone next to where apoptosis was triggered whereas hemichannels also promoted cell death beyond this area. Buffering cytoplasmic Ca(2+) changes inhibited the spread of apoptosis in both cases. We conclude that Cx43 hemichannels, in concert with their GJ counterparts, play a role in communicating cytC-induced apoptotic cell death messages.


Subject(s)
Apoptosis , Connexin 43/biosynthesis , Gap Junctions/metabolism , Glioma/metabolism , Signal Transduction , Animals , Apoptosis/genetics , Calcium/metabolism , Cell Line, Tumor , Connexin 43/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Electroporation , Gap Junctions/genetics , Gene Silencing , Glioma/genetics , Humans , Rats , Signal Transduction/genetics
8.
Glia ; 55(15): 1554-64, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17823969

ABSTRACT

Gliomas are particularly difficult to cure owing largely to their invasive nature. The neoplastic changes of astrocytes which give rise to these tumors frequently include a reduction of connexin43 (Cx43), the most abundant connexin isoform expressed in astrocytes. Cx43 is a subunit of gap junctions (GJ), intercellular channels which directly link the cytosol of adjacent cells and allow the regulated passage of ions and small molecules. To examine the role of Cx43 in glioma motility, we identified two variant C6 cell lines which endogenously express high (C6-H) or low (C6-L) levels of Cx43. In wound healing and transwell assays, C6-H cells were more motile than C6-L cells. To deduce whether Cx43 mediated these differences, assays were conducted on C6-H cells retrovirally transduced with Cx43 shRNA. Coincident with the stable knockdown of endogenous Cx43, a decrease in motility and invasion was observed. Gap junctional intercellular communication was also decreased, however motility assays conducted in the presence of GJ inhibitors did not reveal significant differences in cell motility. C6 cells transfected with full length or C-terminal truncated Cx43 (Cx43DeltaCT) were subjected to the aforementioned motility assays to expose alternate mechanisms of Cx43-mediated motility. Cells expressing full length Cx43 exhibited increased motility while cells expressing Cx43DeltaCT did not. This report, the first in which RNAi has been employed to reduce Cx43 expression in gliomas, indicates that the downregulation of Cx43 decreases motility of C6 cells. Furthermore, it is the first report to suggest that the Cx43 CT plays an important role in glioma motility.


Subject(s)
Brain Neoplasms/pathology , Carcinogens , Connexin 43/chemistry , Connexin 43/physiology , Glioma/pathology , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/physiology , Gap Junctions/drug effects , Immunohistochemistry , Neoplasm Invasiveness/pathology , RNA, Small Interfering/pharmacology , Rats , Structure-Activity Relationship , Wound Healing/physiology
9.
J Comp Neurol ; 440(4): 387-94, 2001 Nov 26.
Article in English | MEDLINE | ID: mdl-11745630

ABSTRACT

Glial-neuronal interactions have been implicated in both normal information processing and neuroprotection. One pathway of cellular interactions involves gap junctional intercellular communication (GJIC). In astrocytes, gap junctions are composed primarily of the channel protein connexin43 (Cx43) and provide a substrate for formation of a functional syncytium implicated in the spatial buffering capacity of astrocytes. To study the function of gap junctions in the brain, we used heterozygous Cx43 null mice, which exhibit reduced Cx43 expression. Western blot analysis showed a reduction in the level of Cx43 protein and GJIC in astrocytes cultured from heterozygote mice. The level of Cx43 is reduced in the adult heterozygote cerebrum to 40% of that present in the wild-type. To assess the effect of reduced Cx43 and GJIC on neuroprotection, we examined brain infarct volume in wild-type and heterozygote mice after focal ischemia. In our model of focal stroke, the middle cerebral artery was occluded at two points, above and below the rhinal fissure. Four days after surgery, mice were killed, the brains were sectioned and analyzed. Cx43 heterozygous null mice exhibited a significantly larger infarct volume compared with wild-type (14.4 +/- 1.4 mm(3) vs. 7.7 +/- 0.82 mm(3), P < 0.002). These results suggest that augmentation of GJIC in astrocytes may contribute to neuroprotection after ischemic injury.


Subject(s)
Connexin 43/genetics , Infarction, Middle Cerebral Artery/pathology , Mice, Transgenic , Stroke/pathology , Animals , Astrocytes/cytology , Cells, Cultured , Female , Gap Junctions/pathology , Gap Junctions/physiology , Heterozygote , Homozygote , Male , Mice
10.
Mol Pathol ; 54(5): 293-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11577170

ABSTRACT

AIMS: To establish whether the ectopic expression of CCN3 (NOV) in glioma cells can interfere with their tumorigenic potential and assess its potential value in molecular medicine. METHODS: Glioma cell lines were used to assess whether differences in the degree of intracellular communication induced by the expression of the gap junction protein connexin 43 (Cx43) is related to the differential expression of CCN3 (NOV). The antiproliferative activity of rat CCN3 (rCCN3; NOV) in glioma cells, has been assessed both in vitro and in vivo with glioma cell lines expressing different amounts of CCN3 (NOV). RESULTS: Upon ectopic expression of Cx43, the growth of C6 glioma cells is decreased. An increase of CCN3 (NOV) expression matches the reduced tumorigenic potential of these transfected cells. The localisation of CCN3 (NOV) is affected by the increased expression of Cx43 in the Cx-13 transfected cells, in which it is detected at areas of cell-cell contact. In a xenograft model, CCN3 (NOV) transfected glioma cells were found to induce tumours to a lesser degree than their parental counterparts, which do not express detectable amounts of CCN3 (NOV). CONCLUSIONS: Previous observations had suggested an inverse relation between CCN3 (NOV) expression in glioma cells and their tumorigenicity. These results establish a direct association between the establishment of functional gap junctional intercellular communication and the expression of rCCN3 (NOV). In addition to a negative effect on murine and human cell growth, CCN3 (NOV) has antiproliferative activity on tumour cells in vivo. Thus, the antiproliferative activity of the CCN3 (NOV) protein might involve reorganisation of cellular contacts that play a crucial role in tumorigenesis. The antiproliferative activity of CCN3 (NOV) established in this work sets the stage for the potential use of CCN proteins in molecular oncology.


Subject(s)
Glioma/pathology , Animals , Cell Communication/physiology , Cell Division/physiology , Connexin 43/physiology , Female , Gene Expression , Humans , Mice , Mice, Nude , Models, Animal , Rats , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured , Up-Regulation
11.
Biochem Cell Biol ; 79(4): 387-98, 2001.
Article in English | MEDLINE | ID: mdl-11527208

ABSTRACT

Recent advances in genetic technology have provided a new platform on which the simultaneous analysis of a large number of genes is possible in a rapid and efficient fashion. To assess the differential expression of human genes during neuronal differentiation, we compared the transcript profiles of undifferentiated, partially differentiated, and fully differentiated NT2/D1 cultures with cDNA expression arrays. Approximately 75 genes (13% of the gene array pool) were differentially expressed during neuronal development of NT2/D1 cells. Genes coding for pyruvate kinase M2 isozyme, clathrin assembly proteins, calmodulin, fibronectin, laminin, thymosin beta-10, and many others were upregulated as NT2/D1 cells differentiated into neurons. In contrast, several kinases, phosphatases, and G-protein coupled receptor genes showed downregulation upon neuronal differentiation. The information provided here is an invaluable reference for characterizing the phenotype of these cells. This information can also be used in cell therapy and transplantation in which the graft microenvironment and interaction with the host tissue is crucial.


Subject(s)
Cell Differentiation/genetics , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Oligonucleotide Array Sequence Analysis , Base Sequence , Cells, Cultured , DNA Primers , DNA, Complementary , Embryo, Mammalian/cytology , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction
12.
Neuroscience ; 105(3): 589-98, 2001.
Article in English | MEDLINE | ID: mdl-11516826

ABSTRACT

Gap junctions, which serve as intercellular channels providing direct cytoplasmic continuity and ionic current flow between adjacent cells, are constituted by connexin proteins. Using an in vitro model of bicuculline-induced epileptiform activity, we asked whether increased connexin levels occur during epileptiform activity in the intact whole hippocampus, freshly isolated from young (15-day-old) mouse brain. Exposure to bicuculline (10 microM), for 2-10 h, induced persistent changes in electrical activities that included enhanced spontaneous field activity (4 h), an epileptiform response to single electrical stimulation (6 h), and spontaneous epileptiform activity (6 h). These electrophysiological changes were not reversed by up to 60 min perfusion with normal artificial cerebrospinal fluid, but were greatly depressed by the gap junction uncoupler, carbenoxolone (120 microM, 10 min). Data from RNase protection assay and immunoblotting showed that among several detected gap junctions, only connexin 32 was affected. After 2-6 h exposure to bicuculline, the connexin 32 mRNA expression was upregulated to 2-3-fold control (P < 0.01), and its protein level was significantly elevated the following 6 h (P < 0.01), at which time electrophysiologically measured evidence of clearly epileptiform activity was apparent. In addition, the transcription factor, c-fos protein, but not the cAMP response element-binding protein, was also found to be increased at the early stage of bicuculline exposure (2 h) compared to control (P < 0.05).Thus, we have found that exposing the acutely isolated hippocampus to bicuculline, induced increased c-fos protein, followed by increased connexin 32 transcript and protein, and concurrently, persistent epileptiform activity that was depressed by carbenoxolone.


Subject(s)
Connexins/metabolism , Epilepsy/metabolism , Gap Junctions/metabolism , Hippocampus/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Up-Regulation/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Ulcer Agents/pharmacology , Bicuculline/pharmacology , Carbenoxolone/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Connexins/drug effects , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Interactions/physiology , Epilepsy/chemically induced , Epilepsy/physiopathology , GABA Antagonists/pharmacology , Gap Junctions/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neurons/drug effects , Organ Culture Techniques , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Up-Regulation/drug effects , Gap Junction beta-1 Protein
13.
Cell Commun Adhes ; 8(4-6): 325-8, 2001.
Article in English | MEDLINE | ID: mdl-12064612

ABSTRACT

Glial-neuronal interactions have been implicated in both normal information processing and neuroprotection. One pathway of cellular interactions involves gap junctional intercellular communication (GJIC). In astrocytes, gap junctions are composed primarily of the channel protein, connexin43 (Cx43), and provide a substrate for formation of a functional syncytium implicated in the process of spatial buffering in the CNS. Thus gap junctional communication may be neuroprotective following a CNS insult that entails glutamate cytotoxicity (i.e. ischemia). We have shown that blocking gap junctions during a glutamate insult to co-cultures of astrocytes and neurons results in increased neuronal injury. To assess the effect of reduced Cx43 and GJIC on neuroprotection, we examined brain infarct volume in wild type and Cx43 heterozygote null mice following focal ischemia. Cx43 heterozygous null mice exhibited a significantly larger infarct volume compared to wild type. At the cellular level, a significant increase in TUNEL positive cells was observed in the penumbral region of the Cx43 heterozygote mice. These results suggest that augmentation of GJIC in astrocytes may contribute to neuroprotection following ischemic injury. These findings support the hypothesis that gap junctions play a neuroprotective role against glutamate cytotoxicity.


Subject(s)
Cell Communication/physiology , Central Nervous System/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Brain Ischemia/metabolism , Carbenoxolone/pharmacology , Cells, Cultured , Coculture Techniques , Connexin 43/genetics , Disease Models, Animal , Glutamic Acid/toxicity , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism
14.
Cell Commun Adhes ; 8(4-6): 441-5, 2001.
Article in English | MEDLINE | ID: mdl-12064632

ABSTRACT

Tumour cells often exhibit erratic cell growth, as well as decreased gap junctional intercellular communication (GJIC). C6 glioma cells are characterized by low levels of gap junction mRNA and protein, and decreased amounts of GJIC when compared with astrocytes. Previous work has shown that C6 glioma cells transfected with connexin43 (C6-Cx43) exhibit decreased proliferation in vivo and in vitro, as well as genes that are differentially expressed between these cells. In this study, RNA levels of two CCN (connective tissue growth factor [CTGF], Cyr61/Cef-10, nephroblastoma overexpressed [NOV]) gene family members are shown to be upregulated in C6-Cx43 cells: Cyr61 and Nov. Cyr61 has previously been shown to increase adhesion, migration and growth in many cell types, whereas NOV has growth suppressive capacities. Cyr61 RNA expression is shown here to respond to serum in quiescent cells in an immediate early gene fashion, independent of Cx43 expression. In contrast, Nov RNA levels remain constant, reflective of transfected Cx43 expression. Furthermore, confocal microscopy indicates that NOV colocalizes with Cx43 plaques at the cell membrane. These findings provide insight into the possible role of Nov and Cyr61 in tumour cells.


Subject(s)
Connexin 43/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Cell Membrane/metabolism , Connective Tissue Growth Factor , Connexin 43/genetics , Culture Media/chemistry , Cysteine-Rich Protein 61 , Gap Junctions/metabolism , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , RNA, Messenger/metabolism , Rats , Tumor Cells, Cultured
15.
Cancer Res ; 60(21): 6018-26, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085522

ABSTRACT

Gap junction expression has been reported to control the growth of a variety of transformed cells. We undertook parallel analysis of connexins Cx32 and Cx43 in glioma cells, which revealed potential mechanisms underlying this phenomenon and led to several novel findings. Cx43, but not Cx32, suppressed C6 glioma cell growth. Paradoxically, Cx32 transfection resulted in severalfold more dye transfer than Cx43. However, Cx43 transfectants shared endogenous metabolites more efficiently than Cx32 transfectants. Interestingly, a significant portion of Cx43 permeants were incorporated into macromolecules more readily than those that transferred via Cx32. Cx43 induced contact inhibition of cell growth but in contrast to other reports, did not affect log phase growth rates. Cell death, senescence, or suppression of growth factor signaling was not involved because no significant alterations were seen in cell viability, telomerase, or mitogen-activated protein kinase activity. However, suppression of cell growth by Cx43 entailed the secretion of growth-regulatory factors. Most notably, a major component of conditioned medium that was affected by Cx43 was found to be MFG-E8 (milk fat globule epidermal growth factor 8), which is involved in cell anchorage and integrin signaling. These results indicate that Cx43 regulates cell growth by the modulation of extracellular growth factors including MFG-E8. Furthermore, the ability of a Cx to regulate cell growth may rely on its ability to mediate the intercellular transfer of endogenous metabolites but not artificial dyes.


Subject(s)
Antigens, Surface , Connexin 43/physiology , Gap Junctions/physiology , Glioma/pathology , Membrane Glycoproteins/antagonists & inhibitors , Milk Proteins , Amino Acid Sequence , Animals , Blotting, Northern , Cell Communication/physiology , Cell Division/physiology , Coloring Agents/pharmacokinetics , Connexin 43/biosynthesis , Connexin 43/genetics , Connexins/biosynthesis , Connexins/genetics , Connexins/physiology , Gap Junctions/metabolism , Glioma/genetics , Glioma/metabolism , Humans , MAP Kinase Signaling System/physiology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Molecular Sequence Data , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , Rats , Telomerase/metabolism , Transfection , Gap Junction beta-1 Protein
16.
J Biol Chem ; 275(44): 34407-14, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10924505

ABSTRACT

Given the roles proposed for gap junctional intercellular communication in neuronal differentiation and growth control, we examined the effects of connexin43 (Cx43) expression in a neuroblastoma cell line. A vesicular stomatitis virus G protein (VSVG)-pseudotyped retrovector was engineered to co-express the green fluorescent protein (GFP) and Cx43 in the communication-deficient neuro-2a (N2a) cell line. The 293 GPG packaging cell line was used to produce VSVG-pseudotyped retrovectors coding for GFP, Cx43, or chimeric Cx43.GFP fusion protein. The titer of viral supernatant, as measured by flow cytometry for GFP fluorescence, was approximately 2.0 x 10(7) colony form units (CFU)/ml and was free of replication-competent retroviruses. After a 7-day treatment with retinoic acid (20 microm), N2a transformants (N2a-Cx43 and N2a-Cx43.GFP) maintained the expression of Cx43 and Cx43.GFP. Expression of both constructs resulted in functional coupling, as evidenced by electrophysiological and dye-injection analysis. Suppression of cell growth correlated with expression of both Cx43 or Cx43.GFP and retinoic acid treatment. Based on morphology and immunocytochemistry for neurofilament, no difference was observed in the differentiation of N2a cells compared with cells expressing Cx43 constructs. In conclusion, constitutive expression of Cx43 in N2a cells does not alter retinoic acid-induced neuronal differentiation but does enhance growth inhibition.


Subject(s)
Cell Differentiation/genetics , Cell Division/genetics , Connexin 43/genetics , Neuroblastoma/pathology , Retroviridae/genetics , Genetic Vectors , Green Fluorescent Proteins , Luminescent Proteins/genetics , Neuroblastoma/genetics , Recombinant Fusion Proteins/genetics , Transfection , Tumor Cells, Cultured , Up-Regulation
17.
Neuroscience ; 98(1): 69-78, 2000.
Article in English | MEDLINE | ID: mdl-10858613

ABSTRACT

We recently characterized the rat brain homolog of mouse muscle CArG-binding protein A initially identified in C2 myogenic cells and showed an inverse temporal correlation between increased expression levels of this messenger RNA, c-fos and zif268 messenger RNA levels following the addition of nerve growth factor to PC12 cells. In addition, we found an inverse correlation between c-Fos protein and CArG-binding protein A messenger RNA levels in the lateral caudate-putamen of rats treated acutely and chronically with the D2 receptor antagonist fluphenazine (phenothiozine typical psychotic). To determine whether D1 receptor stimulation is also capable of inducing CArG-binding protein A up-regulation, drug naive or dopamine-depleted (i.e. 6-hydroxydopamine-lesioned) D1 hypersensitized rats (i.e. rats given repeated daily injections of SKF-82958 for 14days) were acutely injected with the D1 agonist SKF-82958 and examined using a combination of in situ hybridization for CArG binding protein A and immunocytochemistry for c-Fos. Both acutely treated animals and dopamine-depleted hypersensitized animals showed increases in CArG-binding protein A. Moderate increases were found in the medial caudate-putamen and nucleus accumbens core and shell regions following acute treatment whereas large increases in CArG-binding protein A expression levels were found in the medial and lateral caudate-putamen and the shell and core of the nucleus accumbens following hypersensitization. No change in CArG-binding protein A expression level was found in the dopamine-depleted, drug naive animals relative to controls. Regions of the basal ganglia where increases in CArG-binding protein A were detected following each treatment correlated perfectly with c-Fos protein induction. The results demonstrate that CArG-binding protein A responds to SKF-82958 and that the changes in CArG-binding protein A match perfectly with the pattern of c-Fos induction induced by the D1 agonist.


Subject(s)
Benzazepines/pharmacology , DNA-Binding Proteins/genetics , Dopamine Agonists/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Receptors, Dopamine D1/agonists , Repressor Proteins/genetics , Animals , Cell Cycle Proteins , Corpus Striatum/chemistry , Corpus Striatum/physiology , Denervation , Gene Expression/drug effects , Genes, Immediate-Early/physiology , In Situ Hybridization , Male , Oxidopamine , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Ribonucleoproteins , Sympatholytics , Transcription Factors
18.
Brain Res Brain Res Rev ; 32(1): 259-66, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751676

ABSTRACT

Astrocytes are characterized by extensive gap junctional intercellular communication (GJIC) mediated primarily by channels composed of connexin43. In contrast, C6 glioma cells are deficient in connexin expression and gap junctional communication. Transfection of these glioma cells with connexin cDNAs results in changes in cellular phenotype following increased GJIC. Specifically, connexin expression correlates with reduced cellular proliferation and tumorigenicity. To characterize the role of gap junctions in this growth control, we have screened for changes in gene expression by differential display. We have observed that these changes in GJIC are associated with changes in expression of several genes, including those coding for a number of secreted factors which may play a role in modulating the tumor phenotype of these cells. These include the immediate early gene cyr61, ostoepontin and the KC gene (murine homologue of the human gro gene).


Subject(s)
Connexin 43/genetics , Gap Junctions , Gene Expression Regulation, Neoplastic , Glioma , Animals , Blotting, Northern , Cell Communication/genetics , Cell Differentiation/genetics , DNA, Complementary , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Phenotype , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats , Sequence Analysis, DNA , Transfection , Tumor Cells, Cultured
19.
Exp Neurol ; 162(1): 13-26, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10716885

ABSTRACT

Nervous system deficits account for the third largest group of fatal birth defects (after heart and respiratory problems) in North America. Although considerable advance has been made in neuroscience research, the early events involved in neurogenesis remain to be elucidated. More specifically, the effects of signaling molecules on intercellular communication during neurodevelopment have not yet been studied. The development of the central nervous system is regulated, at least in part, by signaling molecules such as bone morphogenetic proteins (BMPs). In this study, we have used the embryonal mouse P19 cell line to examine the effects of BMP2 and BMP4 on gap junctional communication as well as neuronal and astrocytic differentiation. The undifferentiated P19 cells show high levels of the gap junction protein, connexin43 (Cx43), and functional intercellular coupling. However, Cx43 expression and dye coupling decrease as these cells differentiate into neurons and astrocytes. In contrast, cells treated with BMP2 or BMP4 lose their capacity to differentiate into neurons but not astrocytes, while they maintain extensive gap junctional communication. The very few neurons that remain in the BMP-treated cultures are coupled (a characteristic not seen in the control neurons). Together, our data suggest that BMPs may play a critical role in morphogenesis of P19 cells while they affect gap junctions.


Subject(s)
Bone Morphogenetic Proteins/genetics , Gap Junctions/physiology , Neurons/physiology , Transforming Growth Factor beta , Animals , Astrocytes/chemistry , Astrocytes/cytology , Astrocytes/physiology , Blotting, Northern , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Cell Communication/physiology , Cell Differentiation/physiology , Cell Division/physiology , Connexin 43/genetics , Fluoresceins , Fluorescent Dyes , Gap Junctions/chemistry , Gene Expression Regulation, Developmental/physiology , Mice , Neoplastic Stem Cells , Nervous System/cytology , Nervous System/embryology , Neurons/chemistry , Neurons/cytology , RNA, Messenger/analysis , Transcription, Genetic/physiology , Tumor Cells, Cultured
20.
Muscle Nerve Suppl ; 9: S39-48, 2000.
Article in English | MEDLINE | ID: mdl-11135283

ABSTRACT

The X-linked form of Charcot-Marie-Tooth disease has been associated with mutations in the connexin 32 (Cx 32) gene, which encodes a gap junction protein. The majority of identified mutations are missense, but a few nonsense mutations or frame-shifting microdeletions have been encountered. Functional assessments of the mutated gap junction protein have demonstrated altered or simple losses of function. Mutations segregate with a typical clinical phenotype, which is the result of an age-related, progressive neuropathy. The mechanisms that cause the nerve damage are unknown. This report describes the consequences of a unique deletion mutation that eliminates the entire coding sequence of Cx 32, resulting in the absence of the Cx 32 gap junction protein in affected, hemizygous men. The clinical expression of this unique mutation was studied by the clinical, electrophysiological, and pathological evaluation of this kinship of five generations. The resulting severe neuropathy combines features of demyelination, notably in paranodal distribution, and distal accentuated axonal degeneration. The predicted absence of Cx 32 gap junctions is shown to be associated with a severe dysfunction of the axon-Schwann cell unit. Observed changes resemble those of Cx 32-null mice. No central nervous system changes were demonstrated.


Subject(s)
Connexins/genetics , Gap Junctions/chemistry , Gap Junctions/pathology , Gene Deletion , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , X Chromosome , Atrophy , Biopsy , Electrophysiology , Family Health , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Male , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Pedigree , Peroneal Nerve/pathology , Peroneal Nerve/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Gap Junction beta-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...