Network


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

Hotspot


Dive into the research topics where Jessica K. Bernard is active.

Publication


Featured researches published by Jessica K. Bernard.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Specific epidermal growth factor receptor autophosphorylation sites promote mouse colon epithelial cell chemotaxis and restitution

Toshimitsu Yamaoka; Mark R. Frey; Rebecca S. Dise; Jessica K. Bernard; D. Brent Polk

Upon ligand binding, epidermal growth factor (EGF) receptor (R) autophosphorylates on COOH-terminal tyrosines, generating docking sites for signaling partners that stimulate proliferation, restitution, and chemotaxis. Specificity for individual EGFR tyrosines in cellular responses has been hypothesized but not well documented. Here we tested the requirement for particular tyrosines, and associated downstream pathways, in mouse colon epithelial cell chemotactic migration. We compared these requirements to those for the phenotypically distinct restitution (wound healing) migration. Wild-type, Y992/1173F, Y1045F, Y1068F, and Y1086F EGFR constructs were expressed in EGFR(-/-) cells; EGF-induced chemotaxis or restitution were determined by Boyden chamber or modified scratch wound assay, respectively. Pharmacological inhibitors of p38, phospholipase C (PLC), Src, MEK, JNK/SAPK, phosphatidylinositol 3-kinase (PI 3-kinase), and protein kinase C (PKC) were used to block EGF-stimulated signaling. Pathway activation was determined by immunoblot analysis. Unlike wild-type EGFR, Y992/1173F and Y1086F EGFR did not stimulate colon epithelial cell chemotaxis toward EGF; Y1045F and Y1068F EGFR partially stimulated chemotaxis. Only wild-type EGFR promoted colonocyte restitution. Inhibition of p38, PLC, and Src, or Grb2 knockdown, blocked chemotaxis; JNK, PI 3-kinase, and PKC inhibitors or c-Cbl knockdown blocked restitution but not chemotaxis. All four EGFR mutants stimulated downstream signaling in response to EGF, but Y992/1173F EGFR was partially defective in PLCγ activation whereas both Y1068F and Y1086F EGFR failed to activate Src. We conclude that specific EGFR tyrosines play key roles in determining cellular responses to ligand. Chemotaxis and restitution, which have different migration phenotypes and physiological consequences, have overlapping but not identical EGFR signaling requirements.


Journal of Biological Chemistry | 2012

Neuregulin-4 is a survival factor for colon epithelial cells both in culture and in vivo

Jessica K. Bernard; Sean P. McCann; Vrinda Bhardwaj; Mary Kay Washington; Mark R. Frey

Background: Neuregulin-4 is a selective ligand for the ErbB4 receptor tyrosine kinase. Results: In colon epithelial cells, inflammatory cytokine-induced apoptosis is blocked by neuregulin-4, but proliferation and migration are not affected. Conclusion: ErbB4 activation by neuregulin-4 is a specific anti-apoptotic signal. Significance: Neuregulin-4/ErbB4 signaling might be therapeutically useful for diseases involving excessive epithelial apoptosis. Expression of the ErbB4 tyrosine kinase is elevated in colonic epithelial cells during inflammatory bowel disease, whereas ErbB4 overexpression in cultured colonocytes blocks TNF-induced apoptosis in a ligand-dependent manner. Together, these observations suggest that ErbB4 induction may be a protective response. However, the effects of ErbB4 signaling in the colonic epithelium in vivo are not known. Furthermore, previous work on ErbB4 used ligands shared with other receptors, raising the question of whether the observed responses are explicitly due to ErbB4. In this study, we used the ErbB4-specific ligand neuregulin-4 (NRG4) to activate ErbB4 and define its role in colonocyte biology. NRG4 treatment, either in cultured cells or in mice, blocked colonic epithelial apoptosis induced by TNF and IFN-γ. It was also protective in a murine experimental colitis model. NRG4 stimulated phosphorylation of ErbB4 but not other ErbB receptors, indicating that this is a specific response. Furthermore, in contrast to related ligands, NRG4 enhanced cell survival but not proliferation or migration, and stimulated phosphorylation of the anti-apoptotic mediator Akt but not ERK MAPK. Pharmacological inhibition of PI3K/Akt signaling reversed the anti-apoptotic effects of NRG4, confirming the role of this cascade in NRG4-induced cell survival. With regard to the potential clinical importance of this pathway, NRG4 expression was decreased in human inflammatory bowel disease samples and mouse models of colitis, suggesting that activation of ErbB4 is altered in disease. Thus, exogenous NRG4 may be beneficial for disorders in which epithelial apoptosis is part of the pathology.


Carcinogenesis | 2015

ERBB4 is over-expressed in human colon cancer and enhances cellular transformation.

Christopher S. Williams; Jessica K. Bernard; Michelle Demory Beckler; Dana Almohazey; Mary Kay Washington; Jesse J. Smith; Mark R. Frey

The ERBB4 receptor tyrosine kinase promotes colonocyte survival. Herein, we tested whether ERBB4s antiapoptotic signaling promotes transformation and colorectal tumorigenesis. ERBB4 alterations in a The Cancer Genome Atlas colorectal cancer (CRC) data set stratified survival, and in a combined Moffitt Cancer Center and Vanderbilt Medical Center CRC expression data set, ERBB4 message levels were increased at all tumor stages. Similarly, western blot and immunohistochemistry on additional CRC tissue banks showed elevated ERBB4 protein in tumors. ERBB4 was highly expressed in aggressive, dedifferentiated CRC cell lines, and its knockdown in LIM2405 cells reduced anchorage-independent colony formation. In nude mouse xenograft studies, ERBB4 alone was insufficient to induce tumor establishment of non-transformed mouse colonocytes, but its over-expression in cells harboring Apc(min) and v-Ha-Ras caused a doubling of tumor size. ERBB4-expressing xenografts displayed increased activation of survival pathways, including epidermal growth factor receptor and Akt phosphorylation and COX-2 expression, and decreased apoptotic signals. Finally, ERBB4 deletion from mouse intestinal epithelium impaired stem cell replication and in vitro enteroid establishment. In summary, we report that ERBB4 is over-expressed in human CRC, and in experimental systems enhances the survival and growth of cells driven by Ras and/or WNT signaling. Chronic ERBB4 over-expression in the context of, for example, inflammation may contribute to colorectal carcinogenesis. Tumors with high receptor levels are likely to have enhanced cell survival signaling through epidermal growth factor receptor, PI3K and COX-2. These results suggest ERBB4 as a novel therapeutic target in a subset of CRC.


Cell Death and Disease | 2017

ErbB4 signaling stimulates pro-inflammatory macrophage apoptosis and limits colonic inflammation

Michael Schumacher; Matija Hedl; Clara Abraham; Jessica K. Bernard; Patricia R Lozano; Jonathan J. Hsieh; Dana Almohazey; Edie B Bucar; Shivesh Punit; Peter J. Dempsey; Mark R. Frey

Efficient clearance of pro-inflammatory macrophages from tissues after resolution of a challenge is critical to prevent prolonged inflammation. Defects in clearance can contribute to conditions such as inflammatory bowel disease, and thus may be therapeutically targetable. However, the signaling pathways that induce termination of pro-inflammatory macrophages are incompletely defined. We tested whether the ErbB4 receptor tyrosine kinase, previously not known to have role in macrophage biology, is involved in this process. In vitro, pro-inflammatory activation of cultured murine and human macrophages induced ErbB4 expression; in contrast, other ErbB family members were not induced in pro-inflammatory cells, and other innate immune lineages (dendritic cells, neutrophils) did not express detectable ErbB4 levels. Treatment of activated pro-inflammatory macrophages with the ErbB4 ligand neuregulin-4 (NRG4) induced apoptosis. ErbB4 localized to the mitochondria in these cells. Apoptosis was accompanied by loss of mitochondrial membrane potential, and was dependent upon the proteases that generate the cleaved ErbB4 intracellular domain fragment, suggesting a requirement for this fragment and mitochondrial pathway apoptosis. In vivo, ErbB4 was highly expressed on pro-inflammatory macrophages but not neutrophils during experimental DSS colitis in C57Bl/6 mice. Active inflammation in this model suppressed NRG4 expression, which may allow for macrophage persistence and ongoing inflammation. Consistent with this notion, NRG4 levels rebounded during the recovery phase, and administration of exogenous NRG4 during colitis reduced colonic macrophage numbers and ameliorated inflammation. These data define a novel role for ErbB4 in macrophage apoptosis, and outline a mechanism of feedback inhibition that may promote resolution of colitis.


American Journal of Pathology | 2014

The ErbB4 Ligand Neuregulin-4 Protects against Experimental Necrotizing Enterocolitis

Steven J. McElroy; Shannon L. Castle; Jessica K. Bernard; Dana Almohazey; Catherine J. Hunter; Brandon Bell; Denise Al Alam; Larry Wang; Henri R. Ford; Mark R. Frey


The FASEB Journal | 2015

Macrophage-Specific ErbB4 is Induced by DSS Colitis and Regulates Macrophage Survival

Michael Schumacher; Jessica K. Bernard; Shivesh Punit; Jonathan J. Hsieh; Mark R. Frey


Inflammatory Bowel Diseases | 2016

P-151 Neuregulin-4 Induced ErbB4 Signaling in Macrophages Is Protective in DSS Colitis

Michael Schumacher; Jessica K. Bernard; Jon Hsieh; Shivesh Punit; Mark R. Frey


Gastroenterology | 2016

1089 Neuregulin-4 Stimulates Pro-Inflammatory Macrophage Apoptosis Through ADAM17 Dependent Cleavage of ErbB4 to Ameliorate Colitis

Michael Schumacher; Jessica K. Bernard; Jonathan J. Hsieh; Shivesh Punit; Peter J. Dempsey; Mark R. Frey


Gastroenterology | 2015

249 ErbB4 Expression on Macrophages Is Induced by DSS Colitis and Negatively Regulates Macrophage Survival

Michael Schumacher; Jessica K. Bernard; Jonathan J. Hsieh; Mark R. Frey


The FASEB Journal | 2014

ErbB4 deletion compromises the murine small intestinal stem cell niche and sensitizes the epithelium to TNF-induced apoptosis (1119.6)

Dana Almohazey; Jessica K. Bernard; Mark R. Frey

Collaboration


Dive into the Jessica K. Bernard's collaboration.

Top Co-Authors

Avatar

Mark R. Frey

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Dana Almohazey

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Jonathan J. Hsieh

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Shivesh Punit

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Shannon L. Castle

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Catherine J. Hunter

Children's Memorial Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. Brent Polk

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Henri R. Ford

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge