Network


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

Hotspot


Dive into the research topics where Suzanne M. Bernier is active.

Publication


Featured researches published by Suzanne M. Bernier.


Journal of Cellular Physiology | 2003

TNFα suppresses link protein and type II collagen expression in chondrocytes: Role of MEK1/2 and NF-κB signaling pathways

Cheryle A. Séguin; Suzanne M. Bernier

Tumor necrosis factor α (TNFα) inhibits matrix synthesis by chondrocytes in rheumatoid arthritis and osteoarthritis; however, the underlying signaling pathways are poorly characterized. This study investigated the TNFα‐activated pathways regulating expression of two key components of the cartilage matrix—link protein and type II collagen. In rat articular chondrocytes, TNFα decreased link protein and type II collagen mRNA to undetectable levels within 48 h. Levels of link protein mRNA recovered more readily than type II collagen mRNA following removal of the cytokine. TNFα‐mediated reduction in mRNA of both matrix molecules occurred at the level of transcription and, for link protein, mRNA stability. Turnover of type II collagen and link protein mRNA was dependent on new protein synthesis. In both prechondrocytes and articular chondrocytes, TNFα induced concentration‐dependent activation of MEK1/2 and NF‐κB, but not p38 or JNK. Sustained activation of NF‐κB was observed for up to 72 h following continuous or transient exposure to TNFα. Using pharmacological and molecular approaches, the MEK1/2 and NF‐κB pathways were found to mediate inhibition of type II collagen and link protein gene expression by TNFα. Both prechondrocytes and articular chondrocytes are targets of TNFα. This study identifies pathways through which TNFα perturbs the synthesis and organization of articular cartilage matrix during inflammation. J. Cell. Physiol. 197: 357–369, 2003© 2003 Wiley‐Liss, Inc.


Arthritis Research & Therapy | 2008

Forced mobilization accelerates pathogenesis: characterization of a preclinical surgical model of osteoarthritis

C. Thomas G. Appleton; David D. McErlain; V. Pitelka; Neil Schwartz; Suzanne M. Bernier; James L Henry; David W. Holdsworth; Frank Beier

Preclinical osteoarthritis (OA) models are often employed in studies investigating disease-modifying OA drugs (DMOADs). In this study we present a comprehensive, longitudinal evaluation of OA pathogenesis in a rat model of OA, including histologic and biochemical analyses of articular cartilage degradation and assessment of subchondral bone sclerosis. Male Sprague-Dawley rats underwent joint destabilization surgery by anterior cruciate ligament transection and partial medial meniscectomy. The contralateral joint was evaluated as a secondary treatment, and sham surgery was performed in a separate group of animals (controls). Furthermore, the effects of walking on a rotating cylinder (to force mobilization of the joint) on OA pathogenesis were assessed. Destabilization-induced OA was investigated at several time points up to 20 weeks after surgery using Osteoarthritis Research Society International histopathology scores, in vivo micro-computed tomography (CT) volumetric bone mineral density analysis, and biochemical analysis of type II collagen breakdown using the CTX II biomarker. Expression of hypertrophic chondrocyte markers was also assessed in articular cartilage. Cartilage degradation, subchondral changes, and subchondral bone loss were observed as early as 2 weeks after surgery, with considerable correlation to that seen in human OA. We found excellent correlation between histologic changes and micro-CT analysis of underlying bone, which reflected properties of human OA, and identified additional molecular changes that enhance our understanding of OA pathogenesis. Interestingly, forced mobilization exercise accelerated OA progression. Minor OA activity was also observed in the contralateral joint, including proteoglycan loss. Finally, we observed increased chondrocyte hypertrophy during pathogenesis. We conclude that forced mobilization accelerates OA damage in the destabilized joint. This surgical model of OA with forced mobilization is suitable for longitudinal preclinical studies, and it is well adapted for investigation of both early and late stages of OA. The time course of OA progression can be modulated through the use of forced mobilization.


Journal of Leukocyte Biology | 2003

LF15-0195 generates tolerogenic dendritic cells by suppression of NF-κB signaling through inhibition of IKK activity

Jinming Yang; Suzanne M. Bernier; Thomas E. Ichim; Mu Li; Xiaoping Xia; Dejun Zhou; Xuyan Huang; Gill H. Strejan; David J. White; Robert Zhong; Wei-Ping Min

LF15‐0195 (LF) is a potent, less toxic analog of the immunosuppressant 15‐deoxyspergualine, which we previously reported to prevent graft rejection and to induce permanent tolerance in a murine cardiac transplantation model. However, the underlying mechanism of action of LF required elucidation. In this study, dendritic cells (DC) treated with LF before activation with tumor necrosis factor α (TNF‐α)/lipopolysaccharide (LPS) failed to express maturation markers (major histocompatibility complex II, CD40, CD86) and interleukin‐12. LF prevented, in a concentration‐dependent manner, the activation and nuclear translocation of nuclear factor‐κB (NF‐κB) in DC following addition of TNF‐α/LPS. Yet‐activated and active IκB kinases (IKKs) were inhibited in cells pretreated with LF, thereby preventing the phosphorylation of IκB and release of NF‐κB, a key regulator of genes associated with the maturation of DC. LF‐induced inhibition of IKK activity was reversed in a dose‐dependent manner by the overexpression of IKK. The T helper cell type 2 (Th2) differentiation of naïve T cells promoted by LF‐treated DC in vitro correlates with Th2 polarization observed in transplant recipients made tolerant by LF. These data demonstrated that LF‐induced blockade of NF‐κB signaling at the level of IKK promoted the generation of tolerogenic DC that inhibited Th1 polarization and increased Th2 polarization in vitro and in vivo.


Arthritis Research & Therapy | 2006

The induction of CCN2 by TGFβ1 involves Ets-1

Jonathan van Beek; Laura Kennedy; Jason S Rockel; Suzanne M. Bernier; Andrew Leask

CCN2 is encoded by an immediate-early gene induced in mesenchymal cells during the formation of blood vessels, bone and connective tissue. It plays key roles in cell adhesion and migration, as well as matrix remodeling. CCN2 is overexpressed in fibrosis, arthritis and cancer; thus, an understanding of how to control CCN2 expression is likely to have importance in developing therapies to combat these pathologies. Previously, we found that the promoter sequence GAGGAATG is important for Ccn2 gene regulation in NIH 3T3 fibroblasts. In this report, we show that this sequence mediates activation of the CCN2 promoter by the ETS family of transcription factors. Endogenous Ets-1 binds this element of the CCN2 promoter, and dominant negative Ets-1 and specific Ets-1 small interfering RNA block induction of CCN2 expression by TGFβ. In the absence of added TGFβ1, Ets-1, but not the related fli-1, synergizes with Smad 3 to activate the CCN2 promoter. Whereas the ability of transfected Ets-1 to activate the CCN2 promoter is dependent on protein kinase C (PKC), Ets-1 in the presence of co-transfected Smad3 does not require PKC, suggesting that the presence of Smad3 bypasses the requirement of Ets-1 for PKC to activate target promoter activity. Our results are consistent with the notion that Smad3 and Ets-1 cooperate in the induction of the CCN2 promoter by TGFβ1. Antagonizing Ets-1 might be of benefit in attenuating CCN2 expression in fibrosis, arthritis and cancer, and may be useful in modulating the outcome of these disorders.


Cell Communication and Adhesion | 2005

Functional Characterization of Oculodentodigital Dysplasia-Associated Cx43 Mutants

Elizabeth McLachlan; Janet L. Manias; Xiang-Qun Gong; Crystal S. Lounsbury; Qing Shao; Suzanne M. Bernier; Donglin Bai; Dale W. Laird

Oculodentodigital dysplasia (ODDD) is associated with at least 28 connexin43 (Cx43) mutations. We characterized four of these mutants; Q49K, L90V, R202H, and V216L. Populations of these GFP-tagged mutants were transported to the cell surface in Cx43-negative HeLa cells and Cx43-positive NRK cells. Dual patch-clamp functional analysis in N2A cells demonstrated that channels formed by each mutant have dramatically reduced conductance. Dye-coupling analysis revealed that each mutant exhibits a dominant-negative effect on wild-type Cx43. Since ODDD patients display skeletal abnormalities, we examined the effect of three other Cx43 mutants previously shown to exert dominant-negative effects on wild-type Cx43 (G21R, G138R, and G60S) in neonatal calvarial osteoblasts. Differentiation was unaltered by expression of these mutants as alkaline phosphatase activity and extent of culture mineralization were unchanged. This suggests that loss-of-function Cx43 mutants are insufficient to deter committed osteoblasts from their normal function in vitro. Thus, we hypothesize that the bone phenotype of ODDD patients may result from disrupted gap junctional intercellular communication earlier in development or during bone remodeling.


Journal of Bone and Mineral Research | 2008

ODDD‐Linked Cx43 Mutants Reduce Endogenous Cx43 Expression and Function in Osteoblasts and Inhibit Late Stage Differentiation

Elizabeth McLachlan; Isabelle Plante; Qing Shao; Dan Tong; Gerald M. Kidder; Suzanne M. Bernier; Dale W. Laird

Introduction: Bone development and modeling requires precise gap junctional intercellular communication (GJIC). Oculodentodigital dysplasia (ODDD) is an autosomal dominant human disease caused by mutations in the gene (GJA1) encoding the gap junction protein, connexin43 (Cx43). The disease is characterized by craniofacial bone deformities and limb abnormalities. It is our hypothesis that Cx43 mutation causes osteoblast dysfunction, which may contribute to the bone phenotype of ODDD.


Journal of Cellular Physiology | 2007

P2Y nucleotide receptor signaling through MAPK/ERK is regulated by extracellular matrix : Involvement of β3 integrins

Julie C. Kudirka; Nattapon Panupinthu; Mark A. Tesseyman; S. Jeffrey Dixon; Suzanne M. Bernier

Extracellular matrix influences cell behavior through receptors such as integrins and through transmission of mechanical forces. Nucleotides are released in response to mechanical stimuli and bind to P2 nucleotide receptors. As chondrocytes are subjected to frequent mechanical stimulation within a rich extracellular matrix, they are an excellent model for studying integration of signals induced by matrix and nucleotides. We investigated signaling of G protein‐coupled P2Y receptors to MAPK/ERK and how this is influenced by matrix. Rat articular chondrocytes expressed transcripts for P2Y1, P2Y2, P2Y4, and P2Y6 receptors and responded to extracellular nucleotides by transient elevation of cytosolic calcium and MAPK/ERK phosphorylation. ERK1/2 activation was suppressed by the protein kinase C (PKC) inhibitors bisindolylmaleimide I and rottlerin, and by the phospholipase D inhibitor 1‐butanol. Thus, nucleotides stimulate P2Y receptors to activate ERK1/2 through a mechanism dependent on PKC and phospholipase D. We next examined the involvement of integrins. Both an RGD‐containing pentapeptide and a β3 integrin blocking antibody, but not a β1 integrin blocking antibody, abolished nucleotide‐induced ERK1/2 phosphorylation. Moreover, chondrocytes adhering to fibronectin (which binds to β1 and β3 containing integrins in an RGD‐dependent manner) displayed prolonged ERK1/2 signaling compared to cells grown on type I or II collagen (which bind to β1‐containing integrins in an RGD‐independent manner). In conclusion, P2Y receptor signaling through ERK1/2 is gated selectively by matrix proteins. Thus, nucleotides released in response to mechanical stimulation will have differing effects on cell function due to changes in the composition of the extracellular matrix during development and disease. J. Cell. Physiol. 213: 54–64, 2007.


Arthritis Research & Therapy | 2004

Tumor necrosis factor alpha and epidermal growth factor act additively to inhibit matrix gene expression by chondrocyte

Aaron R Klooster; Suzanne M. Bernier

The failure of chondrocytes to replace the lost extracellular matrix contributes to the progression of degenerative disorders of cartilage. Inflammatory mediators present in the joint regulate the breakdown of the established matrix and the synthesis of new extracellular matrix molecules. In the present study, we investigated the effects of tumor necrosis factor alpha (TNF-α) and epidermal growth factor (EGF) on chondrocyte morphology and matrix gene expression. Chondrocytes were isolated from distal femoral condyles of neonatal rats. Cells in primary culture displayed a cobblestone appearance. EGF, but not TNF-α, increased the number of cells exhibiting an elongated morphology. TNF-α potentiated the effect of EGF on chondrocyte morphology. Individually, TNF-α and EGF diminished levels of aggrecan and type II collagen mRNA. In combination, the effects of TNF-α and EGF were additive, indicating the involvement of discrete signaling pathways. Cell viability was not compromised by TNF-α or by EGF, alone or in combination. EGF alone did not activate NF-κB or alter NF-κB activation by TNF-α. Pharmacologic studies indicated that the effects of TNF-α and EGF alone or in combination were independent of protein kinase C signaling, but were dependent on MEK1/2 activity. Finally, we analyzed the involvement of Sox-9 using a reporter construct of the 48 base pair minimal enhancer of type II collagen. TNF-α attenuated enhancer activity as expected; in contrast, EGF did not alter either the effect of TNF-α or basal activity. TNF-α and EGF, acting through distinct signaling pathways, thus have additive adverse effects on chondrocyte function. These findings provide critical insights into the control of chondrocytes through the integration of multiple extracellular signals.


Arthritis Research & Therapy | 2009

Egr-1 inhibits the expression of extracellular matrix genes in chondrocytes by TNFα-induced MEK/ERK signalling

Jason S Rockel; Suzanne M. Bernier; Andrew Leask

IntroductionTNFα is increased in the synovial fluid of patients with rheumatoid arthritis and osteoarthritis. TNFα activates mitogen-activated kinase kinase (MEK)/extracellular regulated kinase (ERK) in chondrocytes; however, the overall functional relevance of MEK/ERK to TNFα-regulated gene expression in chondrocytes is unknown.MethodsChondrocytes were treated with TNFα with or without the MEK1/2 inhibitor U0126 for 24 hours. Microarray analysis and real-time PCR analyses were used to identify genes regulated by TNFα in a MEK1/2-dependent fashion. Promoter/reporter, immunoblot, and electrophoretic mobility shift assays were used to identify transcription factors whose activity in response to TNFα was MEK1/2 dependent. Decoy oligodeoxynucleotides bearing consensus transcription factor binding sites were introduced into chondrocytes to determine the functionality of our results.ResultsApproximately 20% of the genes regulated by TNFα in chondrocytes were sensitive to U0126. Transcript regulation of the cartilage-selective matrix genes Col2a1, Agc1 and Hapln1, and of the matrix metalloproteinase genes Mmp-12 and Mmp-9, were U0126 sensitive – whereas regulation of the inflammatory gene macrophage Csf-1 was U0126 insensitive. TNFα-induced regulation of Sox9 and NFκB activity was also U0126 insensitive. Conversely, TNFα-increased early growth response 1 (Egr-1) DNA binding was U0126 sensitive. Transfection of chondrocytes with cognate Egr-1 oligodeoxynucleotides attenuated the ability of TNFα to suppress Col2a1, Agc1 or Hapln1 mRNA expression.ConclusionsOur results suggest that MEK/ERK and Egr1 are required for TNFα-regulated catabolic and anabolic genes of the cartilage extracellular matrix, and hence may represent potential targets for drug intervention in osteoarthritis or rheumatoid arthritis.


Journal of Cellular Physiology | 2002

Epidermal growth factor stimulates proton efflux from chondrocytic cells

Kevin E.H. Lui; Amiesha S. Panchal; Anu Santhanagopal; S. Jeffrey Dixon; Suzanne M. Bernier

Proton efflux from chondrocytes alters the extracellular pH and ionic composition of cartilage, and influences the synthesis and degradation of extracellular matrix. Epidermal growth factor (EGF) promotes chondrocyte proliferation during skeletal development and accumulates in the synovial fluid in rheumatoid arthritis. The purpose of this study was to investigate the effect of EGF on proton efflux from chondrocytes. When monitored using a Cytosensor microphysiometer, EGF was found to rapidly activate proton efflux from CFK2 chondrocytic cells and rat articular chondrocytes. The actions of EGF were concentration‐dependent with half‐maximal effects at 0.3–0.7 ng/ml. Partial desensitization and time‐dependent recovery of the response were observed following repeated exposures to EGF. EGF‐induced proton efflux was dependent on extracellular glucose, and inhibitors of Na+/H+ exchange (NHE) markedly attenuated the initial increase in proton efflux. The response was diminished by inhibitors of phosphatidylinositol 3‐kinase and phospholipase C, but not by inhibitors of MEK (MAPK/ERK kinase) or protein kinase A or C. Thus, EGF‐induced proton efflux involves glucose metabolism and NHE, and is regulated by a discrete subset of EGF‐activated signaling pathways. In vivo, proton efflux induced by EGF may lead to an acidic environment, enhancing turnover of cartilage matrix during development and in rheumatoid arthritis.

Collaboration


Dive into the Suzanne M. Bernier's collaboration.

Top Co-Authors

Avatar

Frank Beier

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jason S Rockel

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Andrew Leask

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

S. Jeffrey Dixon

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

V. Pitelka

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

C. Thomas G. Appleton

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

David D. McErlain

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

David W. Holdsworth

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Dale W. Laird

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge