Network


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

Hotspot


Dive into the research topics where Cheri R. Owen is active.

Publication


Featured researches published by Cheri R. Owen.


Journal of Neurochemistry | 2001

Brain ischemia and reperfusion activates the eukaryotic initiation factor 2α kinase, PERK

Rita Kumar; Salman Azam; Jonathan M. Sullivan; Cheri R. Owen; Douglas R. Cavener; Peichuan Zhang; David Ron; Heather P. Harding; Jane Jane Chen; Anping Han; Blaine C. White; Gary S. Krause; Donald J. DeGracia

Reperfusion after global brain ischemia results initially in a widespread suppression of protein synthesis in neurons, which persists in vulnerable neurons, that is caused by the inhibition of translation initiation as a result of the phosphorylation of the α‐subunit of eukaryotic initiation factor 2 (eIF2α). To identify kinases responsible for eIF2α phosphorylation [eIF2α(P)] during brain reperfusion, we induced ischemia by bilateral carotid artery occlusion followed by post‐ischemic assessment of brain eIF2α(P) in mice with homozygous functional knockouts in the genes encoding the heme‐regulated eIF2α kinase (HRI), or the amino acid‐regulated eIF2α kinase (GCN2). A 10‐fold increase in eIF2α(P) was observed in reperfused wild‐type mice and in the HRI–/– or GCN2–/– mice. However, in all reperfused groups, the RNA‐dependent protein kinase (PKR)‐like endoplasmic reticulum eIF2α kinase (PERK) exhibited an isoform mobility shift on SDS–PAGE, consistent with the activation of the kinase. These data indicate that neither HRI nor GCN2 are required for the large increase in post‐ischemic brain eIF2α(P), and in conjunction with our previous report that eIF2α(P) is produced in the brain of reperfused PKR–/– mice, provides evidence that PERK is the kinase responsible for eIF2α phosphorylation in the early post‐ischemic brain.


Journal of Cerebral Blood Flow and Metabolism | 2002

Molecular pathways of protein synthesis inhibition during brain reperfusion: Implications for neuronal survival or death

Donald J. DeGracia; Rita Kumar; Cheri R. Owen; Gary S. Krause; Blaine C. White

Protein synthesis inhibition occurs in neurons immediately on reperfusion after ischemia and involves at least alterations in eukaryotic initiation factors 2 (eIF2) and 4 (eIF4). Phosphorylation of the α subunit of eIF2 [eIF2(αP)] by the endoplasmic reticulum transmembrane eIF2α kinase PERK occurs immediately on reperfusion and inhibits translation initiation. PERK activation, along with depletion of endoplasmic reticulum Ca2+ and inhibition of the endoplasmic reticulum Ca2+-ATPase, SERCA2b, indicate that an endoplasmic reticulum unfolded protein response occurs as a consequence of brain ischemia and reperfusion. In mammals, the upstream unfolded protein response components PERK, IRE1, and ATF6 activate prosurvivial mechanisms (e.g., transcription of GRP78, PDI, SERCA2b) and proapoptotic mechanisms (i.e., activation of Jun N-terminal kinases, caspase-12, and CHOP transcription). Sustained eIF2(αP) is proapoptotic by inducing the synthesis of ATF4, the CHOP transcription factor, through “bypass scanning” of 5‘ upstream open-reading frames in ATF4 messenger RNA; these upstream open-reading frames normally inhibit access to the ATF4 coding sequence. Brain ischemia and reperfusion also induce μ-calpain–mediated or caspase-3–mediated proteolysis of eIF4G, which shifts message selection to m7G-cap–independent translation initiation of messenger RNAs containing internal ribosome entry sites. This internal ribosome entry site–mediated translation initiation (i.e., for apoptosis-activating factor-1 and death-associated protein-5) can also promote apoptosis. Thus, alterations in eIF2 and eIF4 have major implications for which messenger RNAs are translated by residual protein synthesis in neurons during brain reperfusion, in turn constraining protein expression of changes in gene transcription induced by ischemia and reperfusion. Therefore, our current understanding shifts the focus from protein synthesis inhibition to the molecular pathways that underlie this inhibition, and the role that these pathways play in prosurvival and proapoptotic processes that may be differentially expressed in vulnerable and resistant regions of the reperfused brain.


Neurological Research | 2001

Endothelial cell activation following moderate traumatic brain injury

Roumen Balabanov; Harold Goldman; Sharon Murphy; Gregory Pellizon; Cheri R. Owen; José A. Rafols; Paula Dore-Duffy

Abstract Traumatic brain injury (TBI) initiates a cascade of acute and chronic injury responses which include disturbances in the cerebrovasculature that may result in the activation of the microvascular endothelial development of a dysfunction endothelium. The present study examines endothelial cell (EC) activation in a percussion model of moderate TBI. The criteria for endothelial activation used in these studies was surface expression of a number of markers collectively termed endothelial activation antigens. Temporal induction of the major histocompatibility (MHC) class II molecules, E-selectin (CD62E), vascular cell adhesion molecule (VACM-1) (CD106) as well as altered expression of constitutively expressed intercellular adhesion molecule-1 (ICAM-1) (CD54), the glucose transporter protein (glut-1), the transferrin receptor (tfR) (CD71), and MHC class I molecules was examined at various times following impact. Induction of E-selectin and increased expression of ICAM-1 was seen by 2 h post-impact (PI) and was sustained through 24 h PI. Decreased expression of immunologically reactive glut-1 and tfR was observed by 2–4 h PI and remained low up to 24 h PI. No induction of VCAM-1, MHC class II molecules or altered constitutive expression or MHC class I molecules was seen. Changes in EC activation were observed predominantly at the site of impact and were diminished temporarily. These results indicate that mild concussive injury to the brain results in activation of the endothelium. [Neurol Res 2001; 23: 175-182]


Journal of Neurochemistry | 2005

PERK is responsible for the increased phosphorylation of eIF2α and the severe inhibition of protein synthesis after transient global brain ischemia

Cheri R. Owen; Rita Kumar; Peichuan Zhang; Barbara C. McGrath; Douglas R. Cavener; Gary S. Krause

Reperfusion after global brain ischemia results initially in a widespread suppression of protein synthesis in neurons that is due to inhibition of translation initiation as a result of the phosphorylation of the α‐subunit of eukaryotic initiation factor 2 (eIF2). To address the role of the eIF2α kinase RNA‐dependent protein kinase‐like endoplasmic reticulum kinase (PERK) in the reperfused brain, transgenic mice with a targeted disruption of the Perk gene were subjected to 20 min of forebrain ischemia followed by 10 min of reperfusion. In wild‐type mice, phosphorylated eIF2α was detected in the non‐ischemic brain and its levels were elevated threefold after 10 min of reperfusion. Conversely, there was no phosphorylated eIF2α detected in the non‐ischemic transgenic mice and there was no sizeable rise in phosphorylated eIF2α levels in the forebrain after ischemia and reperfusion. Moreover, there was a substantial rescue of protein translation in the reperfused transgenic mice. Neither group showed any change in total eIF2α, phosphorylated eukaryotic elongation factor 2 or total eukaryotic elongation factor 2 levels. These data demonstrate that PERK is responsible for the large increase in phosphorylated eIF2α and the suppression of translation early in reperfusion after transient global brain ischemia.


Laboratory Investigation | 2008

Smad3 knockout mice exhibit impaired intestinal mucosal healing

Cheri R. Owen; Lisi Yuan; Marc D. Basson

Altered transforming growth factor-β (TGFβ) expression may contribute to inflammatory bowel disease and modulate epithelial cell restitution. Interference with TGFβ-mediated signaling inhibits excisional skin wound healing, but accelerates healing of incisional cutaneous wounds and wounds in some other tissues. Therefore, we sought to clarify the potential role of Smad3-dependent TGFβ signaling in intestinal mucosal healing in Smad3 null mice. Jejunal serosal application of filter disks saturated with 75% acetic acid yielded a circumscribed reproducible ischemic mucosal ulcer 1 day later. We compared ulcer area at 3 and 5 days to day 1 in Smad3 knockout mice and syngeneic wild-type mice, and evaluated mucosal immunoreactivity at the ulcer edge for TGFβ, phosphorylated (activated) focal adhesion kinase (pFAK), phosphorylated extracellular signal-related kinase (pERK), proliferating cell nuclear antigen and apoptosis by TUNEL. Ulcer healing in Smad3 null mice was 17% less at day 3 (n=14, P=0.022) and 15% less at day 5 (n=14, P=0.004) than in wild-type littermates. In wild-type mice, pFAK, pERK and TGFβ immunoreactivity were elevated in epithelium immediately adjacent to the ulcer compared with more distant mucosa. However, this pattern of immunoreactivity for pFAK, pERK and TGFβ was not observed in Smad3 null mice. Smad3 null mice exhibited increased epithelial proliferation and no differences in apoptotic cell death compared with wild types, suggesting that ulcer healing may reflect differences in restitutive cell migration. Thus, Smad3-dependent disruption of the TGFβ signaling pathway impairs the healing of murine intestinal mucosal ulcers and alters patterns of activated FAK and ERK immunoreactivity important for cell migration at the ulcer edge. These studies suggest a significant role for Smad3-dependent TGFβ signaling in intestinal mucosal healing.


Journal of Clinical Investigation | 2008

Colchicine inhibits pressure-induced tumor cell implantation within surgical wounds and enhances tumor-free survival in mice

David H. Craig; Cheri R. Owen; William C. Conway; Mary F. Walsh; Christina Downey; Marc D. Basson

Iatrogenic tumor cell implantation within surgical wounds can compromise curative cancer surgery. Adhesion of cancer cells, in particular colon cancer cells, is stimulated by exposure to increased extracellular pressure through a cytoskeleton-dependent signaling mechanism requiring FAK, Src, Akt, and paxillin. Mechanical stimuli during tumor resection may therefore negatively impact patient outcome. We hypothesized that perioperative administration of colchicine, which prevents microtubule polymerization, could disrupt pressure-stimulated tumor cell adhesion to surgical wounds and enhance tumor-free survival. Ex vivo treatment of Co26 and Co51 colon cancer cells with colchicine inhibited pressure-stimulated cell adhesion to murine surgical wounds and blocked pressure-induced FAK and Akt phosphorylation. Surgical wound contamination with pressure-activated Co26 and Co51 cells significantly reduced tumor-free survival compared with contamination with tumor cells under ambient pressure. Mice treated with pressure-activated Co26 and Co51 cells from tumors preoperatively treated with colchicine in vivo displayed reduced surgical site implantation and significantly increased tumor-free survival compared with mice exposed to pressure-activated cells from tumors not pretreated with colchicine. Our data suggest that pressure activation of malignant cells promotes tumor development and impairs tumor-free survival and that perioperative colchicine administration or similar interventions may inhibit this effect.


Acta Neuropathologica | 1999

Ultrastructural localization of phosphorylated eIF2α [eIF2α(P)] in rat dorsal hippocampus during reperfusion

Ethan N. Goldstein; Cheri R. Owen; Blaine C. White; José A. Rafols

Abstract During post-ischemic brain reperfusion there is a substantial reduction of protein synthesis in selectively vulnerable neurons. Normal protein synthesis requires a functional translation initiation complex, a key element of which is eukaryotic initiation factor 2 (eIF2), which in a complex with GTP introduces the met-tRNAi. Phosphorylation of Ser51 on the α subunit of eIF2 [eIF2α(P)] generates a competitive inhibitor of eIF2B, thereby preventing the replenishment of GTP onto eIF2, thus blocking translation initiation. It has been shown that the conditional expression of an eIF2α mutant (Asp substituted for Ser51) imitating the negative charge of Ser51 (P) induces apoptosis. During the first 10 min of post-ischemic reperfusion, there is an approximately 20-fold increase in eIF2α(P) seen in the cytoplasm of CA1 hippocampal neurons, and, by 1 h, there is also accumulation of eIF2α(P) in the nucleus. We utilized post-embedding electron microscopical immunogold methods to examine the localization of eIF2α(P) during reperfusion. Immunogold particles (10 nm) were concentrated chiefly along the rough endoplasmic reticulum and in association with the membranes of the nuclear envelope in CA1 neurons. Aggregations of gold particles in the nucleus were concentrated: (1) within and around the nucleolus, (2) associated to strands of heterochromatin, and (3) along putative nuclear filaments. The presence of eIF2α(P) in the nucleolus probably reflects its association with nascent ribosomal subunits. The β-subunit of eIF2 has a zinc finger and polylysine blocks analogous to those on other proteins that affect transcription. The association of eIF2α(P) with chromatin may have important implications for transcription.


American Journal of Surgery | 2008

Supraphysiologic extracellular pressure inhibits intestinal epithelial wound healing independently of luminal nutrient flow

Thomas L. Flanigan; Cheri R. Owen; Christopher P. Gayer; Marc D. Basson

BACKGROUND Luminal pressure may injure the gut mucosa in obstruction, ileus, or inflammatory bowel disease. METHODS We formed Roux-en-Y anastomoses in 19 mice, creating proximal and defunctionalized partially obstructed limbs and a distal limb to vary luminal pressure and flow. We induced mucosal ulcers by serosal acetic acid, and assessed proliferation (proliferating cell nuclear antigen) and ERK (immunoblotting). Parallel studies compared Caco-2 enterocyte migration and proliferation after pressure and/or ERK blockade. RESULTS At 3 days, anastomoses were probe-patent, proximal and distal limbs contained chyme, and defunctionalized limbs were empty. The proximal and defunctionalized limbs showed increased pressure and slower healing despite increased proliferation, ERK protein, and ERK activation. In vitro, pressure decreased Caco-2 migration across collagen or fibronectin, stimulated proliferation, and activated ERK. However, ERK blockade did not prevent pressure effects. CONCLUSIONS Luminal pressure during obstruction or ileus may impair mucosal healing independently of luminal flow despite increased mitosis and ERK activation.


Neurological Research | 2006

Characterization of the eIF2-associated protein p67 during brain ischemia and reperfusion

Cheri R. Owen; Christopher Lipinski; Andrea B. Page; Blaine C. White; Jonathon M. Sullivan; José A. Rafols; Gary S. Krause

Abstract Objectives: Within the first few minutes of reperfusion after global brain ischemia, there is a severe depression of protein translation owing to phosphorylation of the α-subunit of eukaryotic initiation factor 2 (eIF2). There is a 67 kDa peptide (p67) that, in its glycosylated form, binds to eIF2 and protects eIF2α from phosphorylation. Moreover, cells with high p67 content exhibit enhanced resistance to eIF2α phosphorylation. To examine the possibilities that deglycosylation of brain p67 occurs during ischemia and/or early reperfusion or that p67 deglycosylation may be more extensive in the vulnerable neurons, these experiments were undertaken to characterize the localization and activation state of p67 during early brain reperfusion Methods: Western blots using antibodies that recognize total p67, glycosylated p67 and phosphorylated eIF2α were used to characterize total p67 and glycosylated p67 during reperfusion-induced phosphorylation of eIF2α. We also characterized the immunohistochemical distribution of glycosylated p67 before and after brain ischemia and reperfusion. Results: There was a large increase in phosphorylated eIF2α, but there was no decrease in the levels of total or glycosylated p67 from those observed in controls following 10 minutes complete brain ischemia and 10 or 60 minutes subsequent reperfusion. Furthermore, there was no reduction in localized immunostaining for glycosylated p67 in vulnerable neurons during ischemia and reperfusion. Discussion: It does not appear that p67 plays a significant role in regulating the phosphorylation of eIF2α following transient brain ischemia.


Gastroenterology | 2006

The Motogenic Effects of Cyclic Mechanical Strain on Intestinal Epithelial Monolayer Wound Closure Are Matrix Dependent

Jianhu Zhang; Cheri R. Owen; Matthew A. Sanders; Jerrold R. Turner; Marc D. Basson

Collaboration


Dive into the Cheri R. Owen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc D. Basson

University of North Dakota

View shared research outputs
Top Co-Authors

Avatar

Rita Kumar

Wayne State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas R. Cavener

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Peichuan Zhang

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anping Han

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge