Network


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

Hotspot


Dive into the research topics where Kris A. Steinbrecher is active.

Publication


Featured researches published by Kris A. Steinbrecher.


Molecular and Cellular Biology | 2005

Glycogen Synthase Kinase 3β Functions To Specify Gene-Specific, NF-κB-Dependent Transcription

Kris A. Steinbrecher; Willie Wilson; Patricia C. Cogswell; Albert S. Baldwin

ABSTRACT Loss of glycogen synthase kinase 3β (GSK-3β) in mice results in embryonic lethality via hepatocyte apoptosis. Consistent with this result, cells from these mice have diminished nuclear factor κB (NF-κB) activity, implying a functional role for GSK-3β in regulating NF-κB. Here, we have explored mechanisms by which GSK-3β may control NF-κB function. We show that cytokine-induced IκB kinase activity and subsequent phosphorylation of IκBα, p105, and p65 are not affected by the absence of GSK-3β activity. Furthermore, nuclear accumulation of p65 following tumor necrosis factor treatment is unaffected by the loss of GSK-3β. However, NF-κB DNA binding activity is reduced in GSK-3β null cells and in cells treated with a pharmacological inhibitor of GSK-3. Expression of certain NF-κB-regulated genes, such as IκBα and macrophage inflammatory protein 2, is minimally affected by the absence of GSK-3β. Conversely, we have identified a subset of NF-κB-regulated genes, including those for interleukin-6 and monocyte chemoattractant protein 1, that require GSK-3β for efficient expression. We show that efficient localization of p65 to the promoter regions of the interleukin-6 and monocyte chemoattractant protein 1 genes following tumor necrosis factor alpha treatment requires GSK-3β. Therefore, GSK-3β has profound effects on transcription in a gene-specific manner through a mechanism involving control of promoter-specific recruitment of NF-κB.


Nature | 2013

Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection

James M. Ertelt; Jeremy M. Kinder; Tony T. Jiang; Xuzhe Zhang; Lijun Xin; Vandana Chaturvedi; Beverly S. Strong; Joseph E. Qualls; Kris A. Steinbrecher; Theodosia A. Kalfa; Aimen F. Shaaban; Sing Sing Way

Newborn infants are highly susceptible to infection. This defect in host defence has generally been ascribed to the immaturity of neonatal immune cells; however, the degree of hyporesponsiveness is highly variable and depends on the stimulation conditions. These discordant responses illustrate the need for a more unified explanation for why immunity is compromised in neonates. Here we show that physiologically enriched CD71+ erythroid cells in neonatal mice and human cord blood have distinctive immunosuppressive properties. The production of innate immune protective cytokines by adult cells is diminished after transfer to neonatal mice or after co-culture with neonatal splenocytes. Neonatal CD71+ cells express the enzyme arginase-2, and arginase activity is essential for the immunosuppressive properties of these cells because molecular inhibition of this enzyme or supplementation with l-arginine overrides immunosuppression. In addition, the ablation of CD71+ cells in neonatal mice, or the decline in number of these cells as postnatal development progresses parallels the loss of suppression, and restored resistance to the perinatal pathogens Listeria monocytogenes and Escherichia coli. However, CD71+ cell-mediated susceptibility to infection is counterbalanced by CD71+ cell-mediated protection against aberrant immune cell activation in the intestine, where colonization with commensal microorganisms occurs swiftly after parturition. Conversely, circumventing such colonization by using antimicrobials or gnotobiotic germ-free mice overrides these protective benefits. Thus, CD71+ cells quench the excessive inflammation induced by abrupt colonization with commensal microorganisms after parturition. This finding challenges the idea that the susceptibility of neonates to infection reflects immune-cell-intrinsic defects and instead highlights processes that are developmentally more essential and inadvertently mitigate innate immune protection. We anticipate that these results will spark renewed investigation into the need for immunosuppression in neonates, as well as improved strategies for augmenting host defence in this vulnerable population.


Journal of Immunology | 2008

Loss of Epithelial RelA Results in Deregulated Intestinal Proliferative/Apoptotic Homeostasis and Susceptibility to Inflammation

Kris A. Steinbrecher; Eleana Harmel-Laws; Raquel Sitcheran; Albert S. Baldwin

NF-κB plays a central, proinflammatory role in chronic intestinal inflammation, yet recent work suggests a predominantly protective function for this transcription factor group in some cell types of the intestine. We herein describe the conditional deletion of the NF-κB RelA gene in murine intestinal epithelia and determine its function in homeostatic control of enterocyte proliferation/apoptosis and susceptibility to colonic inflammation. Mice lacking RelA in ileal and colonic enterocytes were born in expected Mendelian ratios, and RelA-null epithelia differentiated normally. Spontaneous intestinal disease and death occurred with low penetrance in neonates lacking epithelial RelA. IκBα and IκBβ were significantly diminished in RelA-null epithelia, and endotoxin challenge revealed elevated p50 and c-Rel DNA binding activity as compared with controls. Deletion of RelA resulted in diminished expression of antimicrobial (defensin-related cryptdin 4, defensin-related cryptdin 5, RegIIIγ) and antiapoptotic, prorestitution genes (Bcl-xL, RegIV, IL-11, IL-18), and basal rates of epithelial apoptosis and proliferation were elevated. Mice lacking colonic RelA were sensitive to dextran sodium sulfate-induced colitis. Although experimental colitis enhanced proliferation in cells lacking RelA, sustained epithelial cell apoptosis precluded mucosal healing and decreased animal survival. We conclude that activation of RelA is required for homeostatic regulation of cell death and division in intestinal epithelia, as well as for protection from development of severe, acute inflammation of the intestine.


American Journal of Pathology | 2002

Targeted inactivation of the mouse guanylin gene results in altered dynamics of colonic epithelial proliferation.

Kris A. Steinbrecher; Steve A. Wowk; Jeffrey A. Rudolph; David P. Witte; Mitchell B. Cohen

Heat-stable enterotoxin (STa), elaborated by enterotoxigenic Echerichia coli, is a worldwide cause of secretory diarrhea in infants and travelers. Both STa and guanylin, a peptide structurally similar to STa, increase intracellular cGMP levels after binding to the same intestinal receptor, guanylate cyclase C (GC-C). Distinct from its role as an intestinal secretagogue, guanylin may also have a role in intestinal proliferation, as guanylin expression is lost in intestinal adenomas. To determine the function of guanylin in intestinal epithelia, guanylin null mice were generated using a Cre/loxP-based targeting vector. Guanylin null mice grew normally, were fertile and showed no signs of malabsorption. However, the levels of cGMP in colonic mucosa of guanylin null mice were significantly reduced. The colonic epithelial cell migration rate was increased and increased numbers of colonocytes expressing proliferating cell nuclear antigen (PCNA) were present in crypts of guanylin null mice as well. The apoptotic index was similar in guanylin null mice and littermate controls. We conclude from these studies that loss of guanylin results in increased proliferation of colonic epithelia. We speculate that the increase in colonocyte number is related to decreased levels of cGMP and that this increase in proliferation plays a role in susceptibility to intestinal adenoma formation and/or progression.


Hepatology | 2014

IL-17 signaling accelerates the progression of nonalcoholic fatty liver disease in mice

Isaac T.W. Harley; Traci E. Stankiewicz; Daniel A. Giles; Samir Softic; Leah M. Flick; Monica Cappelletti; Rachel Sheridan; Stavra A. Xanthakos; Kris A. Steinbrecher; R. Balfour Sartor; Rohit Kohli; Christopher L. Karp; Senad Divanovic

Inflammation plays a central pathogenic role in the pernicious metabolic and end‐organ sequelae of obesity. Among these sequelae, nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease in the developed world. The twinned observations that obesity is associated with increased activation of the interleukin (IL)‐17 axis and that this axis can regulate liver damage in diverse contexts prompted us to address the role of IL‐17RA signaling in the progression of NAFLD. We further examined whether microbe‐driven IL‐17A regulated NAFLD development and progression. We show here that IL‐17RA−/− mice respond to high‐fat diet stress with significantly greater weight gain, visceral adiposity, and hepatic steatosis than wild‐type controls. However, obesity‐driven lipid accumulation was uncoupled from its end‐organ consequences in IL‐17RA−/− mice, which exhibited decreased steatohepatitis, nicotinamide adenine dinucleotide phosphate (NADPH)‐oxidase enzyme expression, and hepatocellular damage. Neutralization of IL‐17A significantly reduced obesity‐driven hepatocellular damage in wild‐type mice. Further, colonization of mice with segmented filamentous bacteria (SFB), a commensal that induces IL‐17A production, exacerbated obesity‐induced hepatocellular damage. In contrast, SFB depletion protected from obesity‐induced hepatocellular damage. Conclusion: These data indicate that obesity‐driven activation of the IL‐17 axis is central to the development and progression of NAFLD to steatohepatitis and identify the IL‐17 pathway as a novel therapeutic target in this condition. (Hepatology 2014;59:1830–1839)


Journal of Immunology | 2011

Colonic Eosinophilic Inflammation in Experimental Colitis Is Mediated by Ly6Chigh CCR2+ Inflammatory Monocyte/Macrophage-Derived CCL11

Amanda Waddell; Richard Ahrens; Kris A. Steinbrecher; Burke Donovan; Marc E. Rothenberg; Ariel Munitz; Simon P. Hogan

Recent genome-wide association studies of pediatric inflammatory bowel disease have implicated the 17q12 loci, which contains the eosinophil-specific chemokine gene CCL11, with early-onset inflammatory bowel disease susceptibility. In the current study, we employed a murine model of experimental colitis to define the molecular pathways that regulate CCL11 expression in the chronic intestinal inflammation and pathophysiology of experimental colitis. Bone marrow chimera experiments showed that hematopoietic cell-derived CCL11 is sufficient for CCL11-mediated colonic eosinophilic inflammation. We show that dextran sodium sulfate (DSS) treatment promotes the recruitment of F4/80+CD11b+CCR2+Ly6Chigh inflammatory monocytes into the colon. F4/80+CD11b+CCR2+Ly6Chigh monocytes express CCL11, and their recruitment positively correlated with colonic eosinophilic inflammation. Phenotypic analysis of purified Ly6Chigh intestinal inflammatory macrophages revealed that these cells express both M1- and M2-associated genes, including Il6, Ccl4, Cxcl2, Arg1, Chi3l3, Ccl11, and Il10, respectively. Attenuation of DSS-induced F4/80+CD11b+CCR2+Ly6Chigh monocyte recruitment to the colon in CCR2−/− mice was associated with decreased colonic CCL11 expression, eosinophilic inflammation, and DSS-induced histopathology. These studies identify a mechanism for DSS-induced colonic eosinophilia mediated by Ly6ChighCCR2+ inflammatory monocyte/macrophage-derived CCL11.


PLOS ONE | 2011

Loss of Guanylyl Cyclase C (GCC) Signaling Leads to Dysfunctional Intestinal Barrier

Xiaonan Han; Elizabeth A. Mann; Shila Gilbert; Yanfang Guan; Kris A. Steinbrecher; Marshall H. Montrose; Mitchell B. Cohen

Background Guanylyl Cyclase C (GCC) signaling via uroguanylin (UGN) and guanylin activation is a critical mediator of intestinal fluid homeostasis, intestinal cell proliferation/apoptosis, and tumorigenesis. As a mechanism for some of these effects, we hypothesized that GCC signaling mediates regulation of intestinal barrier function. Methodology/Principal Findings Paracellular permeability of intestinal segments was assessed in wild type (WT) and GCC deficient (GCC−/−) mice with and without lipopolysaccharide (LPS) challenge, as well as in UGN deficient (UGN−/−) mice. IFNγ and myosin light chain kinase (MLCK) levels were determined by real time PCR. Expression of tight junction proteins (TJPs), phosphorylation of myosin II regulatory light chain (MLC), and STAT1 activation were examined in intestinal epithelial cells (IECs) and intestinal mucosa. The permeability of Caco-2 and HT-29 IEC monolayers, grown on Transwell filters was determined in the absence and presence of GCC RNA interference (RNAi). We found that intestinal permeability was increased in GCC−/− and UGN−/− mice compared to WT, accompanied by increased IFNγ levels, MLCK and STAT1 activation in IECs. LPS challenge promotes greater IFNγ and STAT1 activation in IECs of GCC−/− mice compared to WT mice. Claudin-2 and JAM-A expression were reduced in GCC deficient intestine; the level of phosphorylated MLC in IECs was significantly increased in GCC−/− and UGN−/− mice compared to WT. GCC knockdown induced MLC phosphorylation, increased permeability in IEC monolayers under basal conditions, and enhanced TNFα and IFNγ-induced monolayer hyperpermeability. Conclusions/Significance GCC signaling plays a protective role in the integrity of the intestinal mucosal barrier by regulating MLCK activation and TJ disassembly. GCC signaling activation may therefore represent a novel mechanism in maintaining the small bowel barrier in response to injury.


Cancer Research | 2010

Colitis-Associated Cancer Is Dependent on the Interplay between the Hemostatic and Inflammatory Systems and Supported by Integrin αMβ2 Engagement of Fibrinogen

Kris A. Steinbrecher; Netanel A. Horowitz; Elizabeth A. Blevins; Kelley A. Barney; Maureen A. Shaw; Eleana Harmel-Laws; Fred D. Finkelman; Matthew J. Flick; Malinda D. Pinkerton; Kathryn E. Talmage; Keith W. Kombrinck; David P. Witte; Joseph S. Palumbo

A link between colitis and colon cancer is well established, but the mechanisms regulating inflammation in this context are not fully defined. Given substantial evidence that hemostatic system components are powerful modulators of both inflammation and tumor progression, we used gene-targeted mice to directly test the hypothesis that the coagulation factor fibrinogen contributes to colitis-associated colon cancer in mice. This fundamental provisional matrix protein was found to be an important determinant of colon cancer. Fibrinogen deficiency resulted in a dramatic diminution in the number of colonic adenomas formed following azoxymethane/dextran sodium sulfate challenge. More detailed analyses in mice expressing a mutant form of fibrinogen that retains clotting function, but lacks the leukocyte integrin receptor alpha(M)beta(2) binding motif (Fibgamma(390-396A)), revealed that alpha(M)beta(2)-mediated engagement of fibrin(ogen) is mechanistically coupled to local inflammatory processes (e.g., interleukin-6 elaboration) and epithelial alterations that contribute to adenoma formation. Consistent with these findings, the majority of Fibgamma(390-396A) mice developed no discernable adenomas, whereas penetrance was 100% in controls. Furthermore, the adenomas harvested from Fibgamma(390-396A) mice were significantly smaller than those from control mice and less proliferative based on quantitative analyses of mitotic indices, suggesting an additional role for fibrin(ogen) in the growth of established adenomas. These studies show, for the first time, a unique link between fibrin(ogen) and the development of inflammation-driven malignancy. Given the importance of antecedent inflammation in the progression of numerous cancers, these studies suggest that therapies targeting fibrin(ogen)-alpha(M)beta(2) interactions may be useful in preventing and/or treating this important subset of malignancies.


Journal of Immunology | 2010

Loss of T Cell and B Cell Quiescence Precedes the Onset of Microbial Flora-Dependent Wasting Disease and Intestinal Inflammation in Gimap5-Deficient Mice

Michael J. Barnes; Halil Aksoylar; Philippe Krebs; Tristan Bourdeau; Carrie N. Arnold; Yu Xia; Kevin Khovananth; Isaac Engel; Sosathya Sovath; Kristin Lampe; Eleana Laws; Amy Saunders; Geoffrey W. Butcher; Mitchell Kronenberg; Kris A. Steinbrecher; David A. Hildeman; H. Leighton Grimes; Bruce Beutler; Kasper Hoebe

Homeostatic control of the immune system involves mechanisms that ensure the self-tolerance, survival and quiescence of hematopoietic-derived cells. In this study, we demonstrate that the GTPase of immunity associated protein (Gimap)5 regulates these processes in lymphocytes and hematopoietic progenitor cells. As a consequence of a recessive N-ethyl-N-nitrosourea–induced germline mutation in the P-loop of Gimap5, lymphopenia, hepatic extramedullary hematopoiesis, weight loss, and intestinal inflammation occur in homozygous mutant mice. Irradiated fetal liver chimeric mice reconstituted with Gimap5-deficient cells lose weight and become lymphopenic, demonstrating a hematopoietic cell-intrinsic function for Gimap5. Although Gimap5-deficient CD4+ T cells and B cells appear to undergo normal development, they fail to proliferate upon Ag-receptor stimulation although NF-κB, MAP kinase and Akt activation occur normally. In addition, in Gimap5-deficient mice, CD4+ T cells adopt a CD44highCD62LlowCD69low phenotype and show reduced IL-7rα expression, and T-dependent and T-independent B cell responses are abrogated. Thus, Gimap5-deficiency affects a noncanonical signaling pathway required for Ag-receptor–induced proliferation and lymphocyte quiescence. Antibiotic-treatment or the adoptive transfer of Rag-sufficient splenocytes ameliorates intestinal inflammation and weight loss, suggesting that immune responses triggered by microbial flora causes the morbidity in Gimap5-deficient mice. These data establish Gimap5 as a key regulator of hematopoietic integrity and lymphocyte homeostasis.


Embo Molecular Medicine | 2012

Enterocyte STAT5 promotes mucosal wound healing via suppression of myosin light chain kinase-mediated loss of barrier function and inflammation

Shila Gilbert; Rongli Zhang; Lee A. Denson; Richard Moriggl; Kris A. Steinbrecher; Noah F. Shroyer; Xiaonan Han

Epithelial myosin light chain kinase (MLCK)‐dependent barrier dysfunction contributes to the pathogenesis of inflammatory bowel diseases (IBD). We reported that epithelial GM‐CSF–STAT5 signalling is essential for intestinal homeostatic response to gut injury. However, mechanism, redundancy by STAT5 or cell types involved remained foggy. We here generated intestinal epithelial cell (IEC)‐specific STAT5 knockout mice, these mice exhibited a delayed mucosal wound healing and dysfunctional intestinal barrier characterized by elevated levels of NF‐κB activation and MLCK, and a reduction of zonula occludens expression in IECs. Deletion of MLCK restored intestinal barrier function in STAT5 knockout mice, and facilitated mucosal wound healing. Consistently, knockdown of stat5 in IEC monolayers led to increased NF‐κB DNA binding to MLCK promoter, myosin light chain phosphorylation and tight junction (TJ) permeability, which were potentiated by administration of tumour necrosis factor‐α (TNF‐α), and prevented by concurrent NF‐κB knockdown. Collectively, enterocyte STAT5 signalling protects against TJ barrier dysfunction and promotes intestinal mucosal wound healing via an interaction with NF‐κB to suppress MLCK. Targeting IEC STAT5 signalling may be a novel therapeutic approach for treating intestinal barrier dysfunction in IBD.

Collaboration


Dive into the Kris A. Steinbrecher's collaboration.

Top Co-Authors

Avatar

Mitchell B. Cohen

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eleana Harmel-Laws

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Mann

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Simon P. Hogan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

David P. Witte

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lee A. Denson

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph S. Palumbo

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Amanda Waddell

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Flick

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Noah F. Shroyer

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge