Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John F. Bechberger is active.

Publication


Featured researches published by John F. Bechberger.


The Journal of Comparative Neurology | 2001

Connexin43 null mutation increases infarct size after stroke

Ramin Siushansian; John F. Bechberger; David F. Cechetto; Vladimir Hachinski; Christian C. Naus

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 mm3 vs. 7.7 ± 0.82 mm3, P < 0.002). These results suggest that augmentation of GJIC in astrocytes may contribute to neuroprotection after ischemic injury. J. Comp. Neurol. 440:387–394, 2001.


Cancer Research | 2007

Tumor-Suppressive Effects of Pannexin 1 in C6 Glioma Cells

Charles P. Lai; John F. Bechberger; Roger J. Thompson; Brian A. MacVicar; Roberto Bruzzone; Christian C. Naus

Mammalian gap junction proteins, connexins, have long been implicated in tumor suppression. Recently, a novel family of proteins named pannexins has been identified as the mammalian counterpart of the invertebrate gap junction proteins, innexins. To date, pannexin 1 (Panx1) and pannexin 2 (Panx2) mRNAs are reported to be expressed in the brain. Most neoplastic cells, including rat C6 gliomas, exhibit reduced connexin expression, aberrant gap junctional intercellular communication (GJIC), and an increased proliferation rate. When gap junctions are up-regulated by transfecting C6 cells with connexin43, GJIC is restored and the proliferation is reduced. In this study, we examined the tumor-suppressive effects of Panx1 expression in C6 cells. Reverse transcription-PCR analysis revealed that C6 cells do not express any of the pannexin transcripts, whereas its nontumorigenic counterpart, rat primary astrocytes, exhibited mRNAs for all three pannexins. On generation of stable C6 transfectants with tagged Panx1 [myc or enhanced green fluorescent protein (EGFP)], a localization of Panx1 expression to the Golgi apparatus and plasma membrane was observed. In addition, Panx1 transfectants exhibited a flattened morphology, which differs greatly from the spindle-shaped control cells (EGFP only). Moreover, Panx1 expression increased gap junctional coupling as shown by the passage of sulforhodamine 101. Finally, we showed that stable expression of Panx1 in C6 cells significantly reduced cell proliferation in monolayers, cell motility, anchorage-independent growth, and in vivo tumor growth in athymic nude mice. Altogether, we conclude that the loss of pannexin expression may participate in the development of C6 gliomas, whereas restoration of Panx1 plays a tumor-suppressive role.


Journal of Neuroscience Research | 1997

Altered gap junctional communication, intercellular signaling, and growth in cultured astrocytes deficient in connexin43.

Christian C. Naus; John F. Bechberger; Yuchun Zhang; Laurent Venance; Hiroshi Yamasaki; Subhash C. Juneja; Gerald M. Kidder; Christian Giaume

Astrocytes are characterized by extensive intercellular communication mediated primarily by gap junction channels composed of connexin43. To examine this junctional protein in astrocytic functions, astrocytes were cultured from embryonic mice with a null mutation in the connexin43 gene (Reaume et al.: Science 267:1831–1834, 1995). Using anti‐Cx43 antibodies, immunoblotting and immunostaining indicated that homozygous null astrocytes were devoid of Cx43. They are also deficient in intercellular dye transfer. Astrocytes cultured from heterozygous embryos express significantly lower Cx43 compared to wild type, and their dye coupling is reduced. Markers of glial differentiation, such as glial fibrillary acidic protein and S100, appeared similar in all genotypes. Measurement of intercellular calcium concentration following mechanical stimulation of confluent astrocytes revealed that the number of cells affected by a rise in intracellular calcium was reduced in homozygous cultures compared to wild type. In fact, the calcium response in homozygous astrocytes was similar to that observed in wild‐type astrocytes in the presence of a gap junction blocker. The growth rate of astrocytes lacking Cx43 was reduced compared to wild‐type astrocytes. These results suggest that gap junctional intercellular communication mediated by Cx43 is not critical for astrocyte differentiation but is likely involved in the regulation of intercellular calcium signaling and cell growth. J. Neurosci. Res. 49:528–540, 1997.


Experimental Neurology | 1991

Gap junction gene expression in human seizure disorder.

Christian C. Naus; John F. Bechberger; David L. Paul

Increased intercellular coupling has been implicated in contributing to the synchronization of discharges characteristic of epileptic foci. Using RNA blot analysis, the level of mRNA for the heart-type gap junction protein, connexin43, is shown to be elevated in samples of temporal lobe neocortex obtained at the time of surgical resection for intractable seizure disorder. Much lower levels of this mRNA are detectable in peritumoral temporal lobe tissue samples obtained during removal of cerebral tumors. However, in cases where the tumors had induced acute seizures, the level of connexin43 mRNA is higher than that detected in epileptic samples. Similar changes are observed for the mRNA for the liver-type gap junction protein, connexin32, although the observed differences are less dramatic. These findings indicate that there is an increase in the level of connexin mRNA in the temporal cortex of patients exhibiting seizure disorders, suggesting an increase in the synthesis of gap junction protein that may lead to an increase in intercellular coupling.


International Journal of Cancer | 1999

Nitric-oxide production by murine mammary adenocarcinoma cells promotes tumor-cell invasiveness

Amila Orucevic; John F. Bechberger; Angela M. Green; Richard A. Shapiro; Timothy R. Billiar; Peeyush K. Lala

The role of nitric oxide (NO) in tumor biology remains controversial and poorly understood. While a few reports indicate that the presence of NO in tumor cells or their micro‐environment is detrimental for tumor‐cell survival, and consequently their metastatic ability, a large body of data suggests that NO promotes tumor progression. The purpose of this study was to identify the source of NO in the spontaneously metastasizing C3‐L5 murine mammary‐adenocarcinoma model, the role of tumor‐derived NO in tumor‐cell invasiveness, and the mechanisms underlying the invasion‐stimulating effects of tumor‐derived NO. The source of NO was established by immunocytochemical localization of NO synthase (NOS) enzymes in C3‐L5 cells in vitro and transplanted tumors in vivo. An in vitro transwell Matrigel invasion assay was used to test the invasiveness of C3‐L5 cells in the presence or the absence of NO blocking agents or iNOS inducers (IFN‐γ and LPS). The mechanisms underlying the invasion‐stimulating effects of tumor‐derived NO were examined by measuring mRNA expression of matrix metalloproteinases (MMP)‐2 and ‐9, and tissue inhibitors of metalloproteinases (TIMP) 1, 2 and 3 in C3‐L5 cells in various experimental conditions. Results showed that C3‐L5 cells expressed high level of eNOS protein in vitro, and in vivo, both in primary and in metastatic tumors. C3‐L5 cells also expressed iNOS mRNA and protein when cultured in the presence of IFN‐γ and LPS. Constitutively produced NO promoted tumor‐cell invasiveness in vitro by down‐regulating TIMP 2 and TIMP 3. In addition, there was up‐regulation of MMP‐2, when extra NO was induced by IFN‐γ and LPS. In conclusion, NO produced by C3‐L5 cells promoted tumor‐cell invasiveness by altering the balance between MMP‐2 and its inhibitors TIMP‐2 and 3. Thus, our earlier observations of anti‐tumor and anti‐metastatic effects of NO inhibitors in vivo in this tumor model can be explained, at least in part, by reduced tumor‐cell invasiveness. Int. J. Cancer 81:889–896, 1999.


The Journal of Neuroscience | 2009

Involvement of the Cytoplasmic C-Terminal Domain of Connexin43 in Neuronal Migration

Cima Cina; Karen Maass; Martin Theis; Klaus Willecke; John F. Bechberger; Christian C. Naus

During brain development, young neurons closely associate with radial glial while migrating from the ventricular zone (VZ) to the cortical plate (CP) of the neocortex. It has been shown previously that gap junctions are needed for this migration to occur properly, but the precise mechanism responsible is still in question. Here, we used Cre recombinase, driven by the nestin promoter, to conditionally knock-out a floxed coding DNA of the connexin43 (Cx43) gene in mice. Radial glia in the VZ normally express connexin43. They undergo divisions that produce neurons and astrocytes and serve as migratory guides for the daughter cells that they produce. Based on histological analysis, we suggest that removing Cx43 from radial glia alters the normal lamination of the mouse neocortex. To monitor newborn neurons during development, we introduced a plasmid containing green fluorescent protein driven by a neuronal (Tα1 tubulin) promoter into the embryonic neocortex using in utero electroporation. The transfected migrating neurons remain in the VZ/intermediate zone (IZ) of the Cx43 conditional knock-out (Cx43cKO) animals, whereas in Cx43fl/fl mice, neurons migrate through the IZ into the CP, indicating that deletion of Cx43 from nestin-positive cells disrupts neuronal migration. We were able to rescue migration of Cx43cKO neurons by electroporating a cytomegalovirus–Cx43 expression plasmid into the embryonic cortex. In contrast, a C-terminal truncated form of Cx43 failed to rescue neuronal migration. In addition, Cx43K258stop mice, in which Cx43 lacks the last 125 amino acid residues of the cytoplasmic C-terminal domain, gave results similar to those seen with the Cx43cKO mice. This study illustrates that deletion of the C-terminal domain of Cx43 alters neuronal migration in the neocortex.


Neuroscience Letters | 1991

Expression of gap junction genes in astrocytes and C6 glioma cells

Christian C. Naus; John F. Bechberger; Stan Caveney; John X. Wilson

The expression of the gap junction genes coding for the liver-type connexin32 and the heart-type connexin43 was examined in primary cultures of astrocytes and in cultures of C6 glioma cells. In both cell types, only connexin43 mRNA was detectable. However, the level of this mRNA was greatly reduced in C6 glioma cells compared to astrocytes. This was consistent with the further observation that astrocytes in primary culture were extensively dye-coupled, whereas such coupling was very restricted in cultures of C6 glioma cells. Connexin43 was immunocytochemically localized in astrocytes, but was not readily detected in C6 cells.


The Journal of Comparative Neurology | 2007

Expression of connexins in embryonic mouse neocortical development

Cima Cina; John F. Bechberger; Mark A. Ozog; Christian C. Naus

During embryonic development, young neurons migrate from the ventricular zone to the cortical plate of the cerebral cortex. Disturbances in this neuronal migration have been associated with numerous diseases such as mental retardation, double cortex, Down syndrome, and epilepsy. One possible cause of these neuropathologies is an aberration in normal gap junctional communication. At least 20 connexin (Cx) genes encode gap junction proteins in mice and humans. A proper understanding of the role of specific connexins in the developing brain requires the characterization of their spatial and temporal pattern of expression. In the current study we performed all the experiments on mouse developing cortex at embryonic days (E) 14, 16, and 18, timepoints that are highly active with regard to cortical development. Using reverse transcription‐polymerase chain reaction, Western blot analysis, and immunohistochemistry, we found that among the family of gap junction proteins, Cx26, Cx36, Cx37, Cx43, and Cx45 were expressed in the developing cortex of mice, Cx30 and Cx32 were absent, while Cx40 was expressed at a very low level. Our results demonstrate that Cx26 and Cx37 were evenly distributed in the cortical layers of developing brain, while Cx36 and Cx43 were more abundant in the ventricular zone and cortical plate. Cx45 distribution appeared to be more abundant at E18 compared to the other timepoints (E14 and E16). Thus, the present study provides identification and the distribution pattern for Cxs associated with cortical development during normal neuronal migration. J. Comp. Neurol. 504:298–313, 2007.


Frontiers in Cellular Neuroscience | 2014

The connexin43 mimetic peptide Gap19 inhibits hemichannels without altering gap junctional communication in astrocytes

Verónica Abudara; John F. Bechberger; Moises Freitas-Andrade; Marijke De Bock; Nan Wang; Geert Bultynck; Christian C. Naus; Luc Leybaert; Christian Giaume

In the brain, astrocytes represent the cellular population that expresses the highest amount of connexins (Cxs). This family of membrane proteins is the molecular constituent of gap junction channels and hemichannels that provide pathways for direct cytoplasm-to-cytoplasm and inside-out exchange, respectively. Both types of Cx channels are permeable to ions and small signaling molecules allowing astrocytes to establish dynamic interactions with neurons. So far, most pharmacological approaches currently available do not distinguish between these two channel functions, stressing the need to develop new specific molecular tools. In astrocytes two major Cxs are expressed, Cx43 and Cx30, and there is now evidence indicating that at least Cx43 operates as a gap junction channel as well as a hemichannel in these cells. Based on studies in primary cultures as well as in acute hippocampal slices, we report here that Gap19, a nonapeptide derived from the cytoplasmic loop of Cx43, inhibits astroglial Cx43 hemichannels in a dose-dependent manner, without affecting gap junction channels. This peptide, which not only selectively inhibits hemichannels but is also specific for Cx43, can be delivered in vivo in mice as TAT-Gap19, and displays penetration into the brain parenchyma. As a result, Gap19 combined with other tools opens up new avenues to decipher the role of Cx43 hemichannels in interactions between astrocytes and neurons in physiological as well as pathological situations.


Cell Communication and Adhesion | 2001

A Neuroprotective Role for Gap Junctions

Christian C. Naus; Mark A. Ozog; John F. Bechberger; Taizen Nakase

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.

Collaboration


Dive into the John F. Bechberger's collaboration.

Top Co-Authors

Avatar

Christian C. Naus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Nelson K. S. Khoo

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Peeyush K. Lala

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Wun Chey Sin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mark A. Ozog

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Moises Freitas-Andrade

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mahmud Bani-Yaghoub

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Wun-Chey Sin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M.D. Mayan

University of A Coruña

View shared research outputs
Researchain Logo
Decentralizing Knowledge