Network


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

Hotspot


Dive into the research topics where Michael Krzyzaniak is active.

Publication


Featured researches published by Michael Krzyzaniak.


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.


American Journal of Pathology | 2012

Targeting α-7 Nicotinic Acetylcholine Receptor in the Enteric Nervous System: A Cholinergic Agonist Prevents Gut Barrier Failure after Severe Burn Injury

Todd W. Costantini; Michael Krzyzaniak; Gerald Cheadle; James G. Putnam; A. Hageny; Nicole E. Lopez; Brian P. Eliceiri; Vishal Bansal; Raul Coimbra

We have previously shown that vagal nerve stimulation prevents intestinal barrier loss in a model of severe burn injury in which injury was associated with decreased expression and altered localization of intestinal tight junction proteins. α-7 Nicotinic acetylcholine receptor (α-7 nAchR) has been shown to be necessary for the vagus nerve to modulate the systemic inflammatory response, but the role of α-7 nAchR in mediating gut protection remained unknown. We hypothesized that α-7 nAchR would be present in the gastrointestinal tract and that treatment with a pharmacological agonist of α-7 nAchR would protect against burn-induced gut barrier injury. The effects of a pharmacological cholinergic agonist on gut barrier integrity were studied using an intraperitoneal injection of nicotine 30 minutes after injury. Intestinal barrier integrity was examined by measuring permeability to 4-kDa fluorescein isothiocyanate-dextran and by examining changes in expression and localization of the intestinal tight junction proteins occludin and ZO-1. Nicotine injection after injury prevented burn-induced intestinal permeability and limited histological gut injury. Treatment with nicotine prevented decreased expression and altered localization of occludin and ZO-1, as seen in animals undergoing burn alone. Defining the interactions among the vagus nerve, the enteric nervous system, and the intestinal epithelium may lead to development of targeted therapeutics aimed at reducing gut barrier failure and intestinal inflammation after severe injury.


Journal of Neurotrauma | 2012

Ghrelin prevents disruption of the blood-brain barrier after traumatic brain injury.

Nicole E. Lopez; Michael Krzyzaniak; Chelsea Blow; James G. Putnam; Yan T. Ortiz-Pomales; A. Hageny; Brian P. Eliceiri; Raul Coimbra; Vishal Bansal

Significant effort has been focused on reducing neuronal damage from post-traumatic brain injury (TBI) inflammation and blood-brain barrier (BBB)-mediated edema. The orexigenic hormone ghrelin decreases inflammation in sepsis models, and has recently been shown to be neuroprotective following subarachnoid hemorrhage. We hypothesized that ghrelin modulates cerebral vascular permeability and mediates BBB breakdown following TBI. Using a weight-drop model, TBI was created in three groups of mice: sham, TBI, and TBI/ghrelin. The BBB was investigated by examining its permeability to FITC-dextran and through quantification of perivascualar aquaporin-4 (AQP-4). Finally, we immunoblotted for serum S100B as a marker of brain injury. Compared to sham, TBI caused significant histologic neuronal degeneration, increases in vascular permeability, perivascular expression of AQP-4, and serum levels of S100B. Treatment with ghrelin mitigated these effects; after TBI, ghrelin-treated mice had vascular permeability and perivascular AQP-4 and S100B levels that were similar to sham. Our data suggest that ghrelin prevents BBB disruption after TBI. This is evident by a decrease in vascular permeability that is linked to a decrease in AQP-4. This decrease in vascular permeability may diminish post-TBI brain tissue damage was evident by decreased S100B.


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.


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.


Journal of Leukocyte Biology | 2012

Cell surface localization and release of the candidate tumor suppressor Ecrg4 from polymorphonuclear cells and monocytes activate macrophages

Andrew Baird; Raul Coimbra; Xitong Dang; Nicole E. Lopez; Jisook Lee; Michael Krzyzaniak; Robert Winfield; Bruce Potenza; Brian P. Eliceiri

We identified fresh human leukocytes as an abundant source of the candidate epithelial tumor suppressor gene, Ecrg4, an epigenetically regulated gene, which unlike other tumor suppressor genes, encodes an orphan‐secreted, ligand‐like protein. In human cell lines, Ecrg4 gene expression was low, Ecrg4 protein undetectable, and Ecrg4 promoter hypermethylation high (45–90%) and reversible by the methylation inhibitor 5‐AzaC. In contrast, Ecrg4 gene expression in fresh, normal human PBMCs and PMNs was 600–800 times higher than in cultured cell lines, methylation of the Ecrg4 promoter was low (<3%), and protein levels were readily detectable in lysates and on the cell surface. Flow cytometry, immunofluorescent staining, and cell surface biotinylation established that full‐length, 14‐kDa Ecrg4 was localized on PMN and monocyte cell surfaces, establishing that Ecrg4 is a membrane‐anchored protein. LPS treatment induced processing and release of Ecrg4, as detected by flow and immunoblotting, whereas an effect of fMLF treatment on Ecrg4 on the PMN cell surface was detected on the polarized R2 subpopulation of cells. This loss of cell surface Ecrg4 was associated with the detection of intact and processed Ecrg4 in the conditioned media of fresh leukocytes and was shown to be associated with the inflammatory response that follows severe, cutaneous burn injury. Furthermore, incubation of macrophages with a soluble Ecrg4‐derived peptide increased the P‐p65, suggesting that processing of an intact sentinel Ecrg4 on quiescent circulating leukocytes leads to processing from the cell surface following injury and macrophage activation.


Journal of Trauma-injury Infection and Critical Care | 2012

Vagal nerve stimulation decreases blood-brain barrier disruption after traumatic brain injury.

Nicole E. Lopez; Michael Krzyzaniak; Todd W. Costantini; James G. Putnam; A. Hageny; Brian P. Eliceiri; Raul Coimbra; Bansal

BACKGROUND Traumatic brain injury (TBI) may alter sympathetic tone causing autonomic abnormalities and organ dysfunction. Vagal nerve stimulation (VNS) has been shown to decrease inflammation and distant organ injury after TBI. It is unknown whether VNS may reduce blood-brain barrier (BBB) dysfunction after TBI. We hypothesize that VNS prevents TBI-induced breakdown of the BBB, subsequent brain edema, and neuronal injury. METHODS A weight-drop model was used to create severe TBI in balb/c mice. Animals were divided into three groups: TBI—TBI only; TBI or VNS—animals that were treated with 10 minutes of VNS immediately before TBI; and sham—animals with opening of the skull but no TBI and VNS treatment. Brain vascular permeability to injected (Mr 70,000) FITC-dextran was measured by radiated fluorescence 6 hours after injury. Injured tissue sections were stained for perivascular aquaporin 4 (AQP-4), an important protein causing BBB-mediated brain edema. Fluorescence was quantified under laser scanning by confocal microscopy. RESULTS Six hours after TBI, cerebral vascular permeability was increased fourfold compared with sham (mean [SD], 6.6E+08 [5.5E+07] arbitrary fluorescence units [afu] vs. 1.5E+08 [2.9E+07] afu; p < 0.001). VNS prevented the increase in permeability when compared with TBI alone (mean [SD], 3.5 E+08 [8.3E+07] afu vs. 6.6E+08 [5.5E+07] afu; p < 0.05). Perivascular expression of AQP-4 was increased twofold in TBI animals compared with sham (mean [SD], 0.96 [0.12] afu vs. 1.79 [0.37] afu; p < 0.05). Similarly, VNS decreased post-TBI expression of AQP-4 to levels similar to sham (mean [SD], 1.15 [0.12] afu; p < 0.05). CONCLUSION VNS attenuates cerebral vascular permeability and decreases the up-regulation of AQP-4 after TBI. Future studies are needed to assess the mechanisms by which VNS maintains the BBB.


Shock | 2012

Vagal nerve stimulation blocks peritoneal macrophage inflammatory responsiveness after severe burn injury.

Nicole E. Lopez; Michael Krzyzaniak; Todd W. Costantini; Antonio De Maio; Andrew Baird; Brian P. Eliceiri; Raul Coimbra

ABSTRACT Large surface area burn injuries lead to activation of the innate immune system, which can be blocked by parasympathetic inputs mediated by the vagus nerve. We hypothesized that vagal nerve stimulation (VNS) would alter the inflammatory response of peritoneal macrophages after severe burn injury. Male BALB/c mice underwent right cervical VNS before 30% total body surface area steam burn and were compared with animals subjected to burn alone. Peritoneal macrophages were harvested at several time points following injury and exposed to lipopolysaccharide (LPS) in culture conditions. The inflammatory response of peritoneal macrophages was measured by analyzing changes in nuclear factor &kgr;B p65Ser536 phosphorylation using flow cytometry. We found that peritoneal macrophages isolated from mice subjected to burn injury were hyperresponsive to LPS challenge, suggesting burn-induced macrophage activation. We identified a protective role for VNS in blocking peritoneal macrophage activation. Analysis of the phosphorylation state of nuclear factor &kgr;B pathway mediator, p65 Rel A, revealed a VNS-mediated reduction in p65Ser536 phosphorylation levels after exposure to LPS compared with burn alone. In combination, these studies suggest VNS mediates the inflammatory response in peritoneal macrophages by affecting the set point of LPS responsiveness.


Journal of Trauma-injury Infection and Critical Care | 2012

CPSI-121 pharmacologically prevents intestinal barrier dysfunction after cutaneous burn through a vagus nerve-dependent mechanism

Michael Krzyzaniak; Yan T. Ortiz-Pomales; Nicole E. Lopez; Luiz Reys; Gerald Cheadle; Paul L. Wolf; Brian P. Eliceiri; Vishal Bansal; Andrew Baird; Raul Coimbra

Background: We have recently demonstrated the protective effects of electrical stimulation of the vagus nerve in prevention of gut injury after severe burn. Here we evaluate the potential for a pharmacologic agonist of the vagus nerve as an approach to regulate outcomes in preclinical models. We tested a new generation of guanylhydrazone-derived compounds, CPSI-121; a compound that may activate the parasympathetic nervous system through poorly understood mechanisms to determine whether we could prevent intestinal mucosal barrier breakdown. Methods: Male balb/c mice were subjected to a full-thickness, 30% total body surface area steam burn, and the efficacy of CPSI-121 was tested against vagus nerve stimulation (VNS) postburn at 4 hours. Surgical vagotomy was used to disrupt the neuroenteric axis and gut injury prevention was assessed. Gut barrier dysfunction was quantified by permeability to 4-kDa fluorescein isothiocyanate-dextran. Gut injury was assessed by histologic evaluation. Tight junction protein expression (ZO-1 and occludin) was characterized by immunofluorescence and immunoblot. Results: VNS and CPSI-121 administration significantly reduced the permeability to 4-kDa fluorescein isothiocyanate-dextran and maintained normal histology compared with burn. However, abdominal vagotomy eliminated the protective effects of both VNS and CPSI-121. ZO-1 and occludin expression was similar to sham in VNS and CPSI-121-treated burn animals, but significantly altered in burn-vagotomized animals. Splenectomy did not alter the effect of CPSI-121. Conclusion: Similar to direct electrical VNS, CPSI-121 effectively protects the intestinal mucosal barrier from breakdown after severe burn. We suggest that this could represent a noninvasive therapy to prevent end-organ dysfunction after trauma that would be administered during resuscitation.

Collaboration


Dive into the Michael Krzyzaniak'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

A. Hageny

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerald Cheadle

University of California

View shared research outputs
Top Co-Authors

Avatar

Paul L. Wolf

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge