Network


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

Hotspot


Dive into the research topics where Carrie Y. Peterson is active.

Publication


Featured researches published by Carrie Y. Peterson.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Vagal nerve stimulation protects against burn-induced intestinal injury through activation of enteric glia cells

Todd W. Costantini; Vishal Bansal; Michael Krzyzaniak; James G. Putnam; Carrie Y. Peterson; William H. Loomis; Paul L. Wolf; Andrew Baird; Brian P. Eliceiri; Raul Coimbra

The enteric nervous system may have an important role in modulating gastrointestinal barrier response to disease through activation of enteric glia cells. In vitro studies have shown that enteric glia activation improves intestinal epithelial barrier function by altering the expression of tight junction proteins. We hypothesized that severe injury would increase expression of glial fibrillary acidic protein (GFAP), a marker of enteric glial activation. We also sought to define the effects of vagal nerve stimulation on enteric glia activation and intestinal barrier function using a model of systemic injury and local gut mucosal involvement. Mice with 30% total body surface area steam burn were used as model of severe injury. Vagal nerve stimulation was performed to assess the role of parasympathetic signaling on enteric glia activation. In vivo intestinal permeability was measured to assess barrier function. Intestine was collected to investigate changes in histology; GFAP expression was assessed by quantitative PCR, by confocal microscopy, and in GFAP-luciferase transgenic mice. Stimulation of the vagus nerve prevented injury-induced intestinal barrier injury. Intestinal GFAP expression increased at early time points following burn and returned to baseline by 24 h after injury. Vagal nerve stimulation prior to injury increased GFAP expression to a greater degree than burn alone. Gastrointestinal bioluminescence was imaged in GFAP-luciferase transgenic animals following either severe burn or vagal stimulation and confirmed the increased expression of intestinal GFAP. Injection of S-nitrosoglutathione, a signaling molecule released by activated enteric glia cells, following burn exerts protective effects similar to vagal nerve stimulation. Intestinal expression of GFAP increases following severe burn injury. Stimulation of the vagus nerve increases enteric glia activation, which is associated with improved intestinal barrier function. The vagus nerve may mediate the signaling that occurs from the central nervous system to the enteric nervous system following gastrointestinal injury.


Journal of Surgical Research | 2009

Role of p38 MAPK in burn-induced intestinal barrier breakdown.

Todd W. Costantini; Carrie Y. Peterson; Lauren Kroll; William H. Loomis; Brian P. Eliceiri; Andrew Baird; Vishal Bansal; Raul Coimbra

BACKGROUND Severe burn results in intestinal barrier breakdown, which may lead to the generation of a systemic inflammatory response and distant organ injury. Intestinal barrier integrity is regulated, in part, by the tight junction protein myosin light chain kinase (MLCK). Previous studies in cell culture have shown that activation of p38 MAPK plays an important role in modulating intestinal barrier function. We hypothesized that (1) severe burn up-regulates p38 MAPK activation and results in increased intestinal permeability via augmented expression of MLCK, and (2) inhibition of p38 MAPK will prevent the burn-induced increase in MLCK expression, resulting in improved intestinal barrier integrity. MATERIALS AND METHODS Male Balb/c mice were subjected to a 30% total body surface area (TBSA) full thickness steam burn, then randomized to receive an intraperitoneal injection of a p38 MAPK inhibitor (SB203580, 25 mg/kg) or vehicle. In vivo intestinal permeability to 4kDa FITC-Dextran was measured. Expression of phosphorylated p38 MAPK, total p38 MAPK, MLCK, and phosphorylated MLC from intestinal extracts was assessed by immunoblotting. RESULTS Severe burn increased intestinal permeability, which was associated with activation of p38 MAPK, and increased expression of MLCK. Treatment with SB203580 significantly attenuated burn-induced intestinal permeability (212 microg/mL versus 81 microg/mL, P<0.05), and decreased expression of intestinal MLCK resulting in decreased phosphorylation of MLC. CONCLUSION p38 MAPK plays an important role in regulating burn-induced intestinal permeability through activation of MLCK. Inhibition of p38 MAPK may be an important therapeutic target aimed at attenuating intestinal barrier breakdown by preventing the burn-induced alterations in tight junction proteins.


Journal of Trauma-injury Infection and Critical Care | 2010

Efferent vagal nerve stimulation attenuates gut barrier injury after burn: modulation of intestinal occludin expression.

Todd W. Costantini; Vishal Bansal; Carrie Y. Peterson; William H. Loomis; James G. Putnam; Fermin Rankin; Paul L. Wolf; Brian P. Eliceiri; Andrew Baird; Raul Coimbra

INTRODUCTION Severe injury can cause intestinal permeability through decreased expression of tight junction proteins, resulting in systemic inflammation. Activation of the parasympathetic nervous system after shock through vagal nerve stimulation is known to have potent anti-inflammatory effects; however, its effects on modulating intestinal barrier function are not fully understood. We postulated that vagal nerve stimulation improves intestinal barrier integrity after severe burn through an efferent signaling pathway, and is associated with improved expression and localization of the intestinal tight junction protein occludin. METHODS Male balb/c mice underwent right cervical vagal nerve stimulation for 10 minutes immediately before 30% total body surface area, full-thickness steam burn. In a separate arm, animals underwent abdominal vagotomy at the gastroesophageal junction before vagal nerve stimulation and burn. Intestinal barrier injury was assessed by permeability to 4 kDa FITC-dextran, histology, and changes in occludin expression using immunoblotting and confocal microscopy. RESULTS Cervical vagal nerve stimulation decreased burn-induced intestinal permeability to FITC-dextran, returning intestinal permeability to sham levels. Vagal nerve stimulation before burn also improved gut histology and prevented burn-induced changes in occludin protein expression and localization. Abdominal vagotomy abrogated the protective effects of cervical vagal nerve stimulation before burn, resulting in gut permeability, histology, and occludin protein expression similar to burn alone. CONCLUSION Vagal nerve stimulation performed before injury improves intestinal barrier integrity after severe burn through an efferent signaling pathway and is associated with improved tight junction protein expression.


Journal of Trauma-injury Infection and Critical Care | 2010

Stimulating the central nervous system to prevent intestinal dysfunction after traumatic brain injury.

Vishal Bansal; Todd W. Costantini; Seok Yong Ryu; Carrie Y. Peterson; William F. Loomis; James G. Putnam; Brian P. Elicieri; Andrew Baird; Raul Coimbra

BACKGROUND Traumatic brain injury (TBI) causes gastrointestinal dysfunction and increased intestinal permeability. Regulation of the gut barrier may involve the central nervous system. We hypothesize that vagal nerve stimulation prevents an increase in intestinal permeability after TBI. METHODS Balb/c mice underwent a weight drop TBI. Selected mice had electrical stimulation of the cervical vagus nerve before TBI. Intestinal permeability to 4.4 kDa FITC-Dextran was measured 6 hours after injury. Ileum was harvested and intestinal tumor necrosis factor-alpha and glial fibrillary acidic protein (GFAP), a marker of glial activity, were measured. RESULTS TBI increased intestinal permeability compared with sham, 6 hours after injury (98.5 microg/mL +/- 12.5 vs. 29.5 microg/mL +/- 5.9 microg/mL; p < 0.01). Vagal stimulation prevented TBI-induced intestinal permeability (55.8 +/- 4.8 microg/mL vs. 98.49 microg/mL +/- 12.5; p < 0.02). TBI animals had an increase in intestinal tumor necrosis factor-alpha 6 hours after injury compared with vagal stimulation + TBI (45.6 +/- 8.6 pg/mL vs. 24.1 +/- 1.4 pg/mL; p < 0.001). TBI increased intestinal GFAP 6.2-fold higher than sham at 2 hours and 11.5-fold higher at 4 hours after injury (p < 0.05). Intestinal GFAP in vagal stimulation + TBI animals was also 6.7-fold higher than sham at 2 hours, however, intestinal GFAP was 18.0-fold higher at 4 hours compared with sham and 1.6-fold higher than TBI alone (p < 0.05). CONCLUSION In a mouse model of TBI, vagal stimulation prevented TBI-induced intestinal permeability. Furthermore, vagal stimulation increased enteric glial activity and may represent the pathway for central nervous system regulation of intestinal permeability.


Journal of Trauma-injury Infection and Critical Care | 2011

Postinjury vagal nerve stimulation protects against intestinal epithelial barrier breakdown.

Michael Krzyzaniak; Carrie Y. Peterson; William F. Loomis; A. Hageny; Paul L. Wolf; Luiz Reys; James G. Putnam; Brian P. Eliceiri; Andrew Baird; Vishal Bansal; Raul Coimbra

BACKGROUND Vagal nerve stimulation (VNS) can have a marked anti-inflammatory effect. We have previously shown that preinjury VNS prevented intestinal barrier breakdown and preserved epithelial tight junction protein expression. However, a pretreatment model has little clinical relevance for the care of the trauma patient. Therefore, we postulated that VNS conducted postinjury would also have a similar protective effect on maintaining gut epithelial barrier integrity. METHODS Male balb/c mice were subjected to a 30% total body surface area, full-thickness steam burn followed by right cervical VNS at 15, 30, 60, 90, 120, and 150 minutes postinjury. Intestinal barrier dysfunction was quantified by permeability to 4 kDa fluorescein isothiocyanate-Dextran, histologic evaluation, gut tumor necrosis factor-alpha (TNF-α) enzyme-linked immunosorbent assay, and expression of tight junction proteins (myosin light chain kinase, occludin, and ZO-1) using immunoblot and immunoflourescence. RESULTS Histologic examination documented intestinal villi appearance similar to sham if cervical VNS was performed within 90 minutes of burn insult. VNS done after injury decreased intestinal permeability to fluorescein isothiocyanate-Dextran when VNS was ≤90 minutes after injury. Burn injury caused a marked increase in intestinal TNF-α levels. VNS-treated animals had TNF-α levels similar to sham when VNS was performed within 90 minutes of injury. Tight junction protein expression was maintained at near sham values if VNS was performed within 90 minutes of burn, whereas expression was significantly altered in burn. CONCLUSION Postinjury VNS prevents gut epithelial breakdown when performed within 90 minutes of thermal injury. This could represent a therapeutic window and clinically relevant strategy to prevent systemic inflammatory response distant organ injury after trauma.


Journal of Trauma-injury Infection and Critical Care | 2009

Burns, inflammation, and intestinal injury: protective effects of an anti-inflammatory resuscitation strategy.

Todd W. Costantini; Carrie Y. Peterson; Lauren Kroll; William H. Loomis; James G. Putnam; Paul L. Wolf; Brian P. Eliceiri; Andrew Baird; Vishal Bansal; Raul Coimbra

BACKGROUND Intestinal barrier breakdown after severe burn can lead to intestinal inflammation, which may act as the source of the systemic inflammatory response. In vitro intestinal cell studies have shown that mitogen-activated protein kinase (MAPK) signaling is an important modulator of intestinal inflammation. We have previously observed that pentoxifylline (PTX) attenuates burn-induced intestinal permeability and tight junction breakdown. We hypothesized that PTX would limit intestinal barrier breakdown and attenuate inflammatory signaling via the MAPK pathway. METHODS Male balb/c mice underwent 30% total body surface area full-thickness steam burn. Immediately after burn, animals received an intraperitoneal injection of PTX (12.5 mg/kg) in normal saline or normal saline alone. In vivo intestinal permeability to 4 kDa fluorescein isothiocyanate-dextran was measured. Intestinal extracts were obtained to measure interleukin-6 by enzyme-linked immunosorbent assay, and phosphorylated p38 MAPK, p38 MAPK, phosphorylated extracellular signal-related kinase (1/2) (ERK (1/2)), and ERK (1/2) by immunoblotting. Acute lung injury was assessed by histology at 24 hours after burn. RESULTS Administration of PTX immediately after injury attenuated burn-induced intestinal permeability. PTX also decreased the burn-induced phosphorylation of p38 MAPK and decreased phosphorylation of ERK (1/2) at 2 hours and 24 hours after injury. Animals given PTX had decreased intestinal interleukin-6 levels. A single dose of PTX also decreased histologic lung injury at 24 hours after burn. CONCLUSION PTX attenuates burn-induced intestinal permeability and subsequent intestinal inflammation. Use of PTX after burn was also associated with decreased acute lung injury. Because of its compelling anti-inflammatory effects, PTX may be an ideal candidate for use as an immunomodulatory adjunct to resuscitation fluid.


Surgery | 2011

Efferent vagal nerve stimulation attenuates acute lung injury following burn: The importance of the gut-lung axis

Michael Krzyzaniak; Carrie Y. Peterson; Gerald Cheadle; William F. Loomis; Paul L. Wolf; Vince Kennedy; James G. Putnam; Vishal Bansal; Brian P. Eliceiri; Andrew Baird; Raul Coimbra

BACKGROUND The purpose of this study was to assess acute lung injury when protection to the gut mucosal barrier offered by vagus nerve stimulation is eliminated by an abdominal vagotomy. METHODS Male balb/c mice were subjected to 30% total body surface area steam burn with and without electrical stimulation to the right cervical vagus nerve. A cohort of animals were subjected to abdominal vagotomy. Lung histology, myeloperoxidase and ICAM-1 immune staining, myeloperoxidase enzymatic assay, and tissue KC levels were analyzed 24 hours after burn. Additionally, lung IkB-α, NF-kB immunoblots, and NF-kB-DNA binding measured by photon emission analysis using NF-kB-luc transgenic mice were performed. RESULTS Six hours post burn, phosphorylation of both NF-kB p65 and IkB-α were observed. Increased photon emission signal was seen in the lungs of NF-kB-luc transgenic animals. Vagal nerve stimulation blunted NF-kB activation similar to sham animals whereas abdominal vagotomy eliminated the anti-inflammatory effect. After burn, MPO positive cells and ICAM-1 expression in the lung endothelium was increased, and lung histology demonstrated significant injury at 24 hours. Vagal nerve stimulation markedly decreased neutrophil infiltration as demonstrated by MPO immune staining and enzyme activity. Vagal stimulation also markedly attenuated acute lung injury at 24 hours. The protective effects of vagal nerve stimulation were reversed by performing an abdominal vagotomy. CONCLUSION Vagal nerve stimulation is an effective strategy to protect against acute lung injury following burn. Moreover, the protective effects of vagal nerve stimulation in the prevention of acute lung injury are eliminated by performing an abdominal vagotomy. These results establish the importance of the gut-lung axis after burn in the genesis of acute lung injury.


Shock | 2011

Burn-induced acute lung injury requires a functional Toll-like receptor 4.

Michael Krzyzaniak; Gerald Cheadle; Carrie Y. Peterson; William F. Loomis; James G. Putnam; Paul L. Wolf; Andrew Baird; Brian P. Eliceiri; Vishal Bansal; Raul Coimbra

The role of the Toll-like receptor 4 (TLR4), a component of the innate immune system, in the development of burn-induced acute lung injury (ALI) has not been completely defined. Recent data suggested that an intact TLR4 plays a major role in the development of organ injury in sterile inflammation. We hypothesized that burn-induced ALI is a TLR4-dependent process. Male C57BL/6J (TLR4 wild-type [WT]) and C57BL/10ScN (TLR4 knockout [KO]) mice were subjected to a 30% total body surface area steam burn. Animals were killed at 6 and 24 h after the insult. Lung specimens were harvested for histological examination after hematoxylin-eosin staining. In addition, lung myeloperoxidase (MPO) and intercellular adhesion molecule 1 immunostaining was performed. Lung MPO was measured by an enzymatic assay. Total lung keratinocyte-derived chemoattractant (IL-8) content was measured by enzyme-linked immunosorbent assay. Western blot was performed to quantify phosphorylated I&kgr;B&agr;, phosphorylated nuclear factor &kgr;B p65 (NF-&kgr;Bp65), and high mobility group box 1 expression. Acute lung injury, characterized by thickening of the alveolar-capillary membrane, hyaline membrane formation, intraalveolar hemorrhage, and neutrophil infiltration, was seen in WT but not KO animals at 24 h. Myeloperoxidase and intercellular adhesion molecule 1 immunostaining of KO animals was also similar to sham but elevated in WT animals. In addition, a reduction in MPO enzymatic activity was observed in KO mice as well as a reduction in IL-8 levels compared with their WT counterparts. Burn-induced ALI develops within 24 h after the initial thermal insult in our model. Toll-like receptor 4 KO animals were clearly protected and had a much less severe lung injury. Our data suggest that burn-induced ALI is a TLR4-dependent process.


Surgical Infections | 2010

Toll-like receptor-4 mediates intestinal barrier breakdown after thermal injury.

Carrie Y. Peterson; Todd W. Costantini; William H. Loomis; James G. Putnam; Paul L. Wolf; Vishal Bansal; Brian P. Eliceiri; Andrew Baird; Raul Coimbra

OBJECTIVE Toll-like receptor 4 (TLR-4) activation after sterile injury leads to organ dysfunction at distant sites. We have shown previously that intestinal barrier breakdown and alteration of tight junction proteins follows thermal injury; however, the role of TLR-4 in this process remains unclear. We hypothesized that increased intestinal permeability and barrier breakdown after burns is a TLR-4 dependent process; hence, knocking down the TLR-4 gene would have a protective effect on burn-induced intestinal dysfunction. METHODS Male C57BL/6J (TLR-4 wild type [WT]) and C57BL/10ScN (TLR-4 knockout [KO]) mice were assigned randomly to either 30% total body surface area steam burn or sham injury. At 4 h, permeability to intraluminally administered fluorescein isothiocyanate (FITC)-dextran was assessed by measuring the fluorescence of the serum. Intestinal samples were analyzed for the presence of the tight junction protein occludin by immunoblotting and immunohistochemistry. Tumor necrosis factor (TNF)-alpha concentrations in the serum and intestines were measured by enzyme-linked immunosorbent assay at 2 h post-burn. RESULTS Serum concentrations of FITC-dextran were decreased in TLR-4 KO mice compared with TLR-4 WT mice after burn injury (92.0 micrograms/mL and 264.5 micrograms/mL, respectively; p < 0.05). After injury, no difference in intestinal permeability was observed between the TLR-4 KO mice and the TLR-4 WT sham-treated mice. The TLR-4 KO mice had preservation of occludin concentrations after thermal injury in both immunoblot and immunohistochemistry assays, but concentrations were decreased in TLR-4 WT animals. The serum concentrations of TNF-alpha serum were higher in TLR-4 WT burned animals than in the sham-treated mice. The TLR-4 KO animals had unmeasurable concentrations of TNF-alpha. No differences in TNF-alpha were observed in the intestinal tissue at 2 h. CONCLUSIONS Mice with TLR-4 KO have less intestinal permeability to FITC-dextran than do TLR-4 WT mice after burn injury as a result of alterations in the tight junction protein occludin. These findings suggest that the greater intestinal permeability and barrier breakdown after burn injury is a TLR-4-dependent process. Toll-like receptor 4 may provide a useful target for the prevention and treatment of systemic inflammatory response syndrome and multisystem organ failure after injury.


Immunopharmacology and Immunotoxicology | 2010

Pentoxifylline modulates p47phox activation and downregulates neutrophil oxidative burst through PKA-dependent and -independent mechanisms.

Todd W. Costantini; Jessica Deree; Carrie Y. Peterson; James G. Putnam; Tiffany Woon; William H. Loomis; Vishal Bansal; Raul Coimbra

Background and Aim: Pentoxifylline (PTX) has been proven to be an inhibitor of fMLP-induced neutrophil (PMN) oxidative burst and is thought to function by increasing cAMP and Protein kinase A (PKA). We hypothesized that PTX diminishes production of the neutrophil respiratory burst by both PKA-dependent and independent mechanisms. Material and Methods: Human neutrophils were isolated and stimulated with fMLP (1μM) alone or in combination with PTX (2mM). PMN activation was determined by the cytochrome C reduction method in the presence and absence of p38 MAPK (SB203580), ERK (PD98059), and PKA inhibitors (H89). Western blot analysis of Ras, Raf, p38 MAPK, ERK, and Akt was performed in PMNs exposed to fMLP and PTX. Cell membranes were fractionated to measure membrane-associated p47 phox. Treated cells were imaged using confocal microscopy to examine changes in localization of Akt and p47phox. Results: PTX produced a decrease in oxidative burst that was diminished but not abrogated by H89 exposure. The reduction in Ras, Raf, and Akt activation seen with PTX was not effected by the presence of H89. The ability of PTX to attenuate phosphorylation of p38 MAPK and ERK was significantly decreased in the presence of H89, suggesting a PKA-dependent mechanisms. Membrane fractions of neutrophils demonstrate that PTX decreased membrane-associated p47phox, thus diminishing the ability to generate oxidative burst. PTX also decreased membrane localization of Akt and p47phox by confocal microscopy. Conclusions: PTX attenuates activation of signaling molecules involved in activation of p47phox and suppress the subsequent assembly of the NADPH machinery through both PKA-dependent and PKA-independent mechanisms.

Collaboration


Dive into the Carrie Y. Peterson's collaboration.

Top Co-Authors

Avatar

Raul Coimbra

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vishal Bansal

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul L. Wolf

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kirk A. Ludwig

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge