Network


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

Hotspot


Dive into the research topics where Christine B. Kern is active.

Publication


Featured researches published by Christine B. Kern.


Journal of Cellular Biochemistry | 2007

Periostin regulates collagen fibrillogenesis and the biomechanical properties of connective tissues

Russell A. Norris; Brook Damon; Vladimir Mironov; Vladimir Kasyanov; Anand Ramamurthi; Ricardo A. Moreno-Rodriguez; Thomas C. Trusk; Jay D. Potts; Richard L. Goodwin; Jeffrey M. Davis; Stanley Hoffman; Xuejun Wen; Yukiko Sugi; Christine B. Kern; Corey H. Mjaatvedt; Debi Turner; Toru Oka; Simon J. Conway; Jeffery D. Molkentin; Gabor Forgacs; Roger R. Markwald

Periostin is predominantly expressed in collagen‐rich fibrous connective tissues that are subjected to constant mechanical stresses including: heart valves, tendons, perichondrium, cornea, and the periodontal ligament (PDL). Based on these data we hypothesize that periostin can regulate collagen I fibrillogenesis and thereby affect the biomechanical properties of connective tissues. Immunoprecipitation and immunogold transmission electron microscopy experiments demonstrate that periostin is capable of directly interacting with collagen I. To analyze the potential role of periostin in collagen I fibrillogenesis, gene targeted mice were generated. Transmission electron microscopy and morphometric analyses demonstrated reduced collagen fibril diameters in skin dermis of periostin knockout mice, an indication of aberrant collagen I fibrillogenesis. In addition, differential scanning calorimetry (DSC) demonstrated a lower collagen denaturing temperature in periostin knockout mice, reflecting a reduced level of collagen cross‐linking. Functional biomechanical properties of periostin null skin specimens and atrioventricular (AV) valve explant experiments provided direct evidence of the role that periostin plays in regulating the viscoelastic properties of connective tissues. Collectively, these data demonstrate for the first time that periostin can regulate collagen I fibrillogenesis and thereby serves as an important mediator of the biomechanical properties of fibrous connective tissues. J. Cell. Biochem. 101: 695–711, 2007.


Developmental Dynamics | 2008

Origin and fate of cardiac mesenchyme

Brian S. Snarr; Christine B. Kern; Andy Wessels

The development of the embryonic heart is dependent upon the generation and incorporation of different mesenchymal subpopulations that derive from intra‐ and extra‐cardiac sources, including the endocardium, epicardium, neural crest, and second heart field. Each of these populations plays a crucial role in cardiovascular development, in particular in the formation of the valvuloseptal apparatus. Notwithstanding shared mechanisms by which these cells are generated, their fate and function differ profoundly by their originating source. While most of our early insights into the origin and fate of the cardiac mesenchyme has come from experimental studies in avian model systems, recent advances in transgenic mouse technology has enhanced our ability to study these cell populations in the mammalian heart. In this article, we will review the current understanding of the role of cardiac mesenchyme in cardiac morphogenesis and discuss several new paradigms based on recent studies in the mouse. Developmental Dynamics 237:2804–2819, 2008.


Developmental Dynamics | 2006

Proteolytic cleavage of versican during cardiac cushion morphogenesis.

Christine B. Kern; Waleed O. Twal; Corey H. Mjaatvedt; Sarah E. Fairey; Bryan P. Toole; M. Luisa Iruela-Arispe; W. Scott Argraves

The proteoglycan versican is essential to the formation of endocardial cushion mesenchyme by epithelial–mesenchymal transformation (EMT). A potentially important factor in the regulation of versican activity during cushion EMT is proteolysis by ADAMTS metalloproteinases. Using antibodies to the DPEAAE neoepitope created by ADAMTS proteolysis of versican, we detected the amino terminal 70‐kDa versican cleavage fragment in cardiac cushions. Initially (i.e., 9.5 days post coitum [dpc]), the fragment is associated with endocardial cells undergoing EMT and with newly derived mesenchymal cells. ADAMTS‐1 and its cofactor fibulin‐1 were also associated with these cells. As cushions become increasingly populated with mesenchymal cells (10.5–12.5 dpc), the fragment remains asymmetrically distributed compared with the pattern of total versican. Highest levels of the fragment are present in regions immediately subjacent to the endocardium characterized as having densely packed, rounded cells, lacking cellular protrusions. With further development (i.e., 12.5–14.5 dpc), the pattern of fragment distribution within cushions broadens to include the ECM surrounding loosely packed mesenchymal cells in the cushion core. Together, the findings reveal that versican proteolysis leading to the production of the 70‐kDa fragment is integral to the formation and differentiation of endocardial cushion mesenchyme. Developmental Dynamics 235:2238–2247, 2006.


Developmental Dynamics | 2007

Versican Proteolysis Mediates Myocardial Regression During Outflow Tract Development

Christine B. Kern; Russell A. Norris; Robert P. Thompson; W. Scott Argraves; Sarah E. Fairey; Leticia Reyes; Stanley Hoffman; Roger R. Markwald; Corey H. Mjaatvedt

An important phase of cardiac outflow tract (OFT) formation is the remodeling of the distal region of the common outlet in which the myocardial sleeve is replaced by with smooth muscle. Here we demonstrate that expression of the proteoglycan versican is reduced before the loss of myocardium from the distal cardiac outlet concomitant with an increase in production of the N‐terminal cleavage fragment of versican. To test whether versican proteolysis plays a role in OFT remodeling, we determined the effects of adenoviral‐mediated expression of a versican isoform devoid of known matrix metalloproteinase cleavage sites (V3) and an N‐terminal fragment of versican (G1). V3 expression promoted an increase in thickness of the proximal OFT myocardial layer independent of proliferation. In contrast, the G1 domain caused thinning and interruptions of the OFT myocardium. These in vivo findings were consistent with findings using cultured primary cardiomyocytes showing that the V3 promoted myocardial cell–cell association while the G1 domain caused a loss of myocardial cell–cell association. Taken together, we conclude that intact versican and G1‐containing versican cleavage products have opposing effects on myocardial cells and that versican proteolysis may facilitate the loss of distal myocardium during OFT remodeling. Developmental Dynamics 236:671–683, 2007.


Developmental Biology | 2008

Fibulin-1 is required for morphogenesis of neural crest-derived structures

Marion A. Cooley; Christine B. Kern; Victor M. Fresco; Andy Wessels; Robert P. Thompson; Tim C. McQuinn; Waleed O. Twal; Corey H. Mjaatvedt; Christopher J. Drake; W. Scott Argraves

Here we report that mouse embryos homozygous for a gene trap insertion in the fibulin-1 (Fbln1) gene are deficient in Fbln1 and exhibit cardiac ventricular wall thinning and ventricular septal defects with double outlet right ventricle or overriding aorta. Fbln1 nulls also display anomalies of aortic arch arteries, hypoplasia of the thymus and thyroid, underdeveloped skull bones, malformations of cranial nerves and hemorrhagic blood vessels in the head and neck. The spectrum of malformations is consistent with Fbln1 influencing neural crest cell (NCC)-dependent development of these tissues. This is supported by evidence that Fbln1 expression is associated with streams of cranial NCCs migrating adjacent to rhombomeres 2-7 and that Fbln1-deficient embryos display patterning anomalies of NCCs forming cranial nerves IX and X, which derive from rhombomeres 6 and 7. Additionally, Fbln1-deficient embryos show increased apoptosis in areas populated by NCCs derived from rhombomeres 4, 6 and 7. Based on these findings, it is concluded that Fbln1 is required for the directed migration and survival of cranial NCCs contributing to the development of pharyngeal glands, craniofacial skeleton, cranial nerves, aortic arch arteries, cardiac outflow tract and cephalic blood vessels.


Journal of Molecular and Cellular Cardiology | 2010

β-Adrenergic Receptor Stimulated Ncx1 Upregulation is Mediated via a CaMKII/AP-1 Signaling Pathway in Adult Cardiomyocytes

Santhosh K. Mani; Erin A. Egan; Benjamin Addy; Michael Grimm; Harinath Kasiganesan; Thirumagal Thiyagarajan; Ludivine Renaud; Joan Heller Brown; Christine B. Kern; Donald R. Menick

The Na(+)-Ca(2+) exchanger gene (Ncx1) is upregulated in hypertrophy and is often found elevated in end-stage heart failure. Studies have shown that the change in its expression contributes to contractile dysfunction. beta-Adrenergic receptor (beta-AR) signaling plays an important role in the regulation of calcium homeostasis in the cardiomyocyte, but chronic activation in periods of cardiac stress contributes to heart failure by mechanisms which include Ncx1 upregulation. Here, using a Ca(2+)/calmodulin-dependent protein kinase II (CaMKIIdelta(c)) null mouse, we demonstrate that beta-AR-stimulated Ncx1 upregulation is dependent on CaMKII. beta-AR-stimulated Ncx1 expression is mediated by activator protein 1 (AP-1) factors and is independent of cAMP-response element-binding protein (CREB) activation. The MAP kinases (ERK1/2, JNK and p38) are not required for AP-1 factor activation. Chromatin immunoprecipitation demonstrates that beta-AR stimulation activates the ordered recruitment of JunB homodimers, which then are replaced by c-Jun homodimers binding to the proximal AP-1 elements of the endogenous Ncx1 promoter. In conclusion, this work has provided insight into the intracellular signaling pathways and transcription factors regulating Ncx1 gene expression in a chronically beta-AR-stimulated heart.


Journal of Molecular and Cellular Cardiology | 2013

Insufficient versican cleavage and Smad2 phosphorylation results in bicuspid aortic and pulmonary valves

Loren E. Dupuis; Hanna Osinska; Michael Weinstein; Robert B. Hinton; Christine B. Kern

Bicuspid or bifoliate aortic valve (BAV) results in two rather than three cusps and occurs in 1-2% of the population placing them at higher risk of developing progressive aortic valve disease. Only NOTCH-1 has been linked to human BAV, and genetically modified mouse models of BAV are limited by low penetrance and additional malformations. Here we report that in the Adamts5(-/-) valves, collagen I, collagen III, and elastin were disrupted in the malformed hinge region that anchors the mature semilunar cusps and where the ADAMTS5 proteoglycan substrate versican, accumulates. ADAMTS5 deficient prevalvular mesenchyme also exhibited a reduction of α-smooth muscle actin and filamin A suggesting versican cleavage may be involved in TGFβ signaling. Subsequent evaluation showed a significant decrease of pSmad2 in regions of prevalvular mesenchyme in Adamts5(-/-) valves. To test the hypothesis that ADAMTS5 versican cleavage is required, in part, to elicit Smad2 phosphorylation we further reduced Smad2 in Adamts5(-/-) mice through intergenetic cross. The Adamts5(-/-);Smad2(+/-) mice had highly penetrant BAV and bicuspid pulmonary valve (BPV) malformations as well as increased cusp and hinge size compared to the Adamts5(-/-) and control littermates. These studies demonstrate that semilunar cusp malformations (BAV and BPV) can arise from a failure to remodel the proteoglycan-rich provisional ECM. Specifically, faulty versican clearance due to ADAMTS5 deficiency blocks the initiation of pSmad2 signaling, which is required for excavation of endocardial cushions during aortic and pulmonary valve development. Further studies using the Adamts5(-/-); Smad2(+/-) mice with highly penetrant and isolated BAV, may lead to new pharmacological treatments for valve disease.


Anatomy and Embryology | 2005

Detection of βig-H3, a TGFβ induced gene, during cardiac development and its complementary pattern with periostin

Russell A. Norris; Christine B. Kern; Andy Wessels; Elaine E. Wirrig; Roger R. Markwald; Corey H. Mjaatvedt

Regulation of normal cardiac development involves numerous transcription factors, cytoskeletal proteins, signaling molecules, and extracellular matrix proteins. These key molecular components act in concert to induce morphological changes essential for the proper development of a functional four-chambered heart. Growth factors such as BMPs and TGFβ’s play a role in migration, proliferation and differentiation during cardiac development and are important regulators of the extracellular matrix (ECM). Genes responsive to these morphogens are likely to play an equally significant role during cardiac development. Therefore, we sought to clone the chicken TGFβ induced gene βig-H3 and evaluate its spatio-temporal expression during heart morphogenesis. Our studies show by Northern analysis, whole mount and section in situ hybridization experiments that βig-H3 is expressed primarily in the mesenchyme of the atrioventricular and outflow tract cushions and later in the right and left atrioventricular valve leaflets and supporting valve structures. The mRNA expression domains of βig-H3 show a complementary pattern compared to that of its highly homologous relative, periostin.


The FASEB Journal | 2009

Histone deacetylases facilitate sodium/calcium exchanger up-regulation in adult cardiomyocytes

Sangeetha Chandrasekaran; Richard E. Peterson; Santhosh K. Mani; Benjamin Addy; Avery L. Buchholz; Lin Xu; Thirumagal Thiyagarajan; Harinath Kasiganesan; Christine B. Kern; Donald R. Menick

It is becoming increasingly evident that histone deacetylases (HDACs) have a prominent role in the alteration of gene expression during the growth remodeling process of cardiac hypertrophy. HDACs are generally viewed as corepressors of gene expression. However, we demonstrate that class I and class II HDACs play an important role in the basal expression and up‐regulation of the sodium calcium exchanger (Ncx1) gene in adult cardiomyocytes. Treatment with the HDAC inhibitor trichostatin A (TSA) prevented the pressure‐overload‐stimulated up‐regulation of Ncxl expression. Overexpression of HDAC5 resulted in the dose‐dependent up‐regulation of basal and a‐adrenergic stimulated Ncx1 expression. We show that Nkx2.5 recruits HDAC5 to the Ncx1 promoter, where HDAC5 complexes with HDAC1. Nkx2.5 also interacts with transcriptional activator p300, which is recruited to the Ncxl promoter. We demonstrate that when Nkx2.5 is acetylated, it is found associated with HDAC5, whereas deacetylated Nkx2.5 is in complex with p300. Notably, TSA treatment prevents p300 from being recruited to the endogenous Ncx1 promoter, resulting in the repression of Ncx1 expression. We propose a novel model for Ncx1 regulation in which deacetylation of Nkx2.5 is required for the recruitment of p300 and results in up‐regulation of exchanger expression.—Chandrasekaran, S., Peterson, R. E., Mani, S. K., Addy, B., Buchholz, A. L., Xu, L., Thiyagarajan, T., Kasiganesan, H., Kern, C. B., Menick, D. R. Histone deacetylases facilitate sodium/calcium exchanger up‐regulation in adult cardiomyocytes. FASEBJ. 23, 3851–3864 (2009). www.fasebj.org


Matrix Biology | 2012

Maladaptive Matrix Remodeling and Regional Biomechanical Dysfunction in a Mouse Model of Aortic Valve Disease

Varun K. Krishnamurthy; Amy Opoka; Christine B. Kern; Farshid Guilak; Daria A. Narmoneva; Robert B. Hinton

Aortic valve disease (AVD) occurs in 2.5% of the general population and often requires surgical intervention. Aortic valve malformation (AVM) underlies the majority of cases, suggesting a developmental etiology. Elastin haploinsufficiency results in complex cardiovascular problems, and 20-45% of patients have AVM and/or AVD. Elastin insufficient (Eln+/-) mice demonstrate AVM and latent AVD due to abnormalities in the valve annulus region. The objective of this study was to examine extracellular matrix (ECM) remodeling and biomechanical properties in regional aortic valve tissue and determine the impact of early AVM on late AVD in the Eln+/- mouse model. Aortic valve ECM composition and remodeling from juvenile, adult, and aged stages were evaluated in Eln+/- mice using histology, ELISA, immunohistochemistry and gelatin zymography. Aortic valve tissue biomechanical properties were determined using micropipette aspiration. Cartilage-like nodules were demonstrated within the valve annulus region at all stages identifying a developmental abnormality preceding AVD. Interestingly, maladaptive ECM remodeling was observed in early AVM without AVD and worsened with late AVD, as evidenced by increased MMP-2 and MMP-9 expression and activity, as well as abnormalities in ADAMTS-mediated versican processing. Cleaved versican was increased in the valve annulus region of aged Eln+/- mice, and this abnormality correlated temporally with adverse alterations in valve tissue biomechanical properties and the manifestation of AVD. These findings identify maladaptive ECM remodeling in functional AVM as an early disease process with a progressive natural history, similar to that seen in human AVD, emphasizing the importance of the annulus region in pathogenesis. Combining molecular and engineering approaches provides complementary mechanistic insights that may be informative in the search for new therapeutic targets and durable valve bioprostheses.

Collaboration


Dive into the Christine B. Kern's collaboration.

Top Co-Authors

Avatar

Corey H. Mjaatvedt

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Andy Wessels

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Donald R. Menick

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Loren E. Dupuis

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

W. Scott Argraves

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Benjamin Addy

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Santhosh K. Mani

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Roger R. Markwald

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Russell A. Norris

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Harinath Kasiganesan

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge