Network


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

Hotspot


Dive into the research topics where Laura Brudecki is active.

Publication


Featured researches published by Laura Brudecki.


Infection and Immunity | 2012

Myeloid-derived suppressor cells evolve during sepsis and can enhance or attenuate the systemic inflammatory response.

Laura Brudecki; Donald A. Ferguson; Charles E. McCall; Mohamed El Gazzar

ABSTRACT Myeloid-derived suppressor cells (MDSCs) are a heterogeneous Gr1+ CD11b+ population of immature cells containing granulocytic and monocytic progenitors, which expand under nearly all inflammatory conditions and are potent repressors of T-cell responses. Studies of MDSCs during inflammatory responses, including sepsis, suggest they can protect or injure. Here, we investigated MDSCs during early and late sepsis. To do this, we used our published murine model of cecal ligation and puncture (CLP)-induced polymicrobial sepsis, which transitions from an early proinflammatory phase to a late anti-inflammatory and immunosuppressive phase. We confirmed that Gr1+ CD11b+ MDSCs gradually increase after CLP, reaching ∼88% of the bone marrow myeloid series in late sepsis. Adoptive transfer of early (day 3) MDSCs from septic mice into naive mice after CLP increased proinflammatory cytokine production, decreased peritoneal bacterial growth, and increased early mortality. Conversely, transfer of late (day 12) MDSCs from septic mice had the opposite effects. Early and late MDSCs studied ex vivo also differed in their inflammatory phenotypes. Early MDSCs expressed nitric oxide and proinflammatory cytokines, whereas late MDSCs expressed arginase activity and anti-inflammatory interleukin 10 (IL-10) and transforming growth factor β (TGF-β). Late MDSCs had more immature CD31+ myeloid progenitors and, when treated ex vivo with granulocyte-macrophage colony-stimulating factor (GM-CSF), generated fewer macrophages and dendritic cells than early MDSCs. We conclude that as the sepsis inflammatory process progresses, the heterogeneous MDSCs shift to a more immature state and from being proinflammatory to anti-inflammatory.


Infection and Immunity | 2014

MicroRNA 21 (miR-21) and miR-181b Couple with NFI-A To Generate Myeloid-Derived Suppressor Cells and Promote Immunosuppression in Late Sepsis

Clara McClure; Laura Brudecki; Donald A. Ferguson; Zhi Q. Yao; Jonathan P. Moorman; Charles E. McCall; Mohamed El Gazzar

ABSTRACT The sepsis initial hyperinflammatory reaction, if not treated early, shifts to a protracted state of immunosuppression that alters both innate and adaptive immunity and is associated with elevated mortality. Myeloid-derived suppressor cells (MDSCs) are myeloid progenitors and precursors that fail to differentiate into mature innate-immunity cells and are known for their potent immunosuppressive activities. We previously reported that murine MDSCs expand dramatically in the bone marrow during late sepsis, induced by cecal ligation and puncture, and demonstrated that they contribute to late-sepsis immunosuppression. However, the molecular mechanism responsible for generating these immature Gr1+ CD11b+ myeloid cells during sepsis remains unknown. We show here that sepsis generates a microRNA (miRNA) signature that expands MDSCs. We found that miRNA 21 (miR-21) and miR-181b expression is upregulated in early sepsis and sustained in late sepsis. Importantly, we found that simultaneous in vivo blockade of both miRNAs via antagomiR (a chemically modified miRNA inhibitor) injection after sepsis initiation decreased the bone marrow Gr1+ CD11b+ myeloid progenitors, improved bacterial clearance, and reduced late-sepsis mortality by 74%. Gr1+ CD11b+ cells isolated from mice injected with antagomiRs were able to differentiate ex vivo into macrophages and dendritic cells and produced smaller amounts of the immunosuppressive interleukin 10 (IL-10) and transforming growth factor β (TGF-β) after stimulation with bacterial lipopolysaccharide, suggesting that immature myeloid cells regained their maturation potential and have lost their immunosuppressive activity. In addition, we found that the protein level of transcription factor NFI-A, which plays a role in myeloid cell differentiation, was increased during sepsis and that antagomiR injection reduced its expression. Moreover, knockdown of NFI-A in the Gr1+ CD11b+ cells isolated from late-septic mice increased their maturation potential and reduced their production of the immunosuppressive mediators, similar to antagomiR injection. These data support the hypothesis that sepsis reprograms myeloid cells and thus alters the innate immunity cell repertoire to promote immunosuppression, and they demonstrate that this process can be reversed by targeting miR-21 and miR-181b to improve late-sepsis survival.


Infection and Immunity | 2012

Hematopoietic Stem-Progenitor Cells Restore Immunoreactivity and Improve Survival in Late Sepsis

Laura Brudecki; Donald A. Ferguson; Deling Yin; Gene LeSage; Charles E. McCall; Mohamed El Gazzar

ABSTRACT Sepsis progresses from an early/acute hyperinflammatory to a late/chronic hypoinflammatory phase with immunosuppression. As a result of this phenotypic switch, mortality in late sepsis from persistent primary infection or opportunistic new infection often exceeds that in acute sepsis. Emerging data support that persistence of the hypoinflammatory (hyporesponsive) effector immune cells during late sepsis might involve alterations in myeloid differentiation/maturation that generate circulating repressor macrophages that do not readily clear active infection. Here, we used a cecal ligation and puncture (CLP) murine model of prolonged sepsis to show that adoptive transfer of CD34+ hematopoietic stem-progenitor cells after CLP improves long-term survival by 65%. CD34+ cell transfer corrected the immunosuppression of late sepsis by (i) producing significantly higher levels of proinflammatory mediators upon ex vivo stimulation with the Toll-like receptor 4 (TLR4) agonist lipopolysaccharide, (ii) enhancing phagocytic activity of peritoneal macrophages, and (iii) clearing bacterial peritonitis. Improved immunity by CD34+ cell transfer decreased inflammatory peritoneal exudate of surviving late-sepsis mice. Cell tracking experiments showed that the transferred CD34+ cells first appeared in the bone marrow and then homed to the spleen and peritoneum. Because CD34+ cells did not affect the early-phase hyperinflammatory response, it is likely that the newly incorporated pluripotent CD34+ cells differentiated into competent immune cells in blood and tissue, thereby reversing or replacing the hyporesponsive endotoxin-tolerant cells that occur and persist after the initiation of early sepsis.


Immunology and Cell Biology | 2013

MicroRNA-146a and RBM4 form a negative feed-forward loop that disrupts cytokine mRNA translation following TLR4 responses in human THP-1 monocytes

Laura Brudecki; Donald A. Ferguson; Charles E. McCall; Mohamed El Gazzar

Within hours after its initiation, the severe systemic inflammatory response of sepsis shifts to an adaptive anti‐inflammatory state with coincident immunosuppression. This anti‐inflammatory phenotype is characterized by diminished proinflammatory cytokine gene expression in response to toll‐like receptor (TLR) stimulation with bacterial endotoxin/lipopolysaccharide (LPS), also known as endotoxin tolerance/adaptation. Our and other studies have established that gene‐specific reprogramming following TLR4 responses independently represses transcription and translation of proinflammatory genes such as tumor necrosis factor alpha (TNFα). We also previously demonstrated that TNFα and interleukin (IL)‐6 mRNA translation is repressed in endotoxin‐adapted THP‐1 human monocytes by an miRNA‐based mechanism involving the argonaute family protein argonaute 2 (Ago2). Here, we further define the molecular nature of reprogramming translation by showing that TLR4‐induced microRNA‐146 promotes a feed‐forward loop that modifies the subcellular localization of the RNA‐binding protein RBM4 (RNA‐binding motif protein 4) and promotes its interaction with Ago2. This interaction results in the assembly of a translation‐repressor complex that disrupts TNFα and IL‐6 cytokine synthesis in endotoxin‐adapted THP‐1 monocytes. This novel molecular path prevents the phosphorylation of RBM4 on serine‐309 by p38 MAPK (mitogen‐activated protein kinase), which leads to RBM4 accumulation in the cytosol and interaction with Ago2. We further find that microRNA‐146a knockdown by antagomirs or protein phosphatase inhibition by okadaic acid increases p38 MAPK phosphorylation and results in RBM4 serine‐309 phosphorylation and nuclear relocalization, which disrupts RBM4 and Ago2 interactions and restores TLR4‐dependent synthesis of TNFα and IL‐6. We conclude that miR‐146a has a diverse and critical role in limiting an excessive acute inflammatory reaction.


Journal of Biological Chemistry | 2009

Role of α-Subunit VISIT-DG Sequence Residues Ser-347 and Gly-351 in the Catalytic Sites of Escherichia coli ATP Synthase

Wenzong Li; Laura Brudecki; Alan E. Senior; Zulfiqar Ahmad

This paper describes the role of α-subunit VISIT-DG sequence residues αSer-347 and αGly-351 in catalytic sites of Escherichia coli F1Fo ATP synthase. X-ray structures show the very highly conserved α-subunit VISIT-DG sequence in close proximity to the conserved phosphate-binding residues αArg-376, βArg-182, βLys-155, and βArg-246 in the phosphate-binding subdomain. Mutations αS347Q and αG351Q caused loss of oxidative phosphorylation and reduced ATPase activity of F1Fo in membranes by 100- and 150-fold, respectively, whereas αS347A mutation showed only a 13-fold loss of activity and also retained some oxidative phosphorylation activity. The ATPase of αS347Q mutant was not inhibited, and the αS347A mutant was slightly inhibited by MgADP-azide, MgADP-fluoroaluminate, or MgADP-fluoroscandium, in contrast to wild type and αG351Q mutant. Whereas 7-chloro-4-nitrobenzo-2-oxa-1, 3-diazole (NBD-Cl) inhibited wild type and αG351Q mutant ATPase essentially completely, ATPase in αS347A or αS347Q mutant was inhibited maximally by ∼80–90%, although reaction still occurred at residue βTyr-297, proximal to the α-subunit VISIT-DG sequence, near the phosphate-binding pocket. Inhibition characteristics supported the conclusion that NBD-Cl reacts inβE (empty) catalytic sites, as shown previously by x-ray structure analysis. Phosphate protected against NBD-Cl inhibition in wild type and αG351Q mutant but not in αS347Q or αS347A mutant. The results demonstrate that αSer-347 is an additional residue involved in phosphate-binding and transition state stabilization in ATP synthase catalytic sites. In contrast, αGly-351, although strongly conserved and clearly important for function, appears not to play a direct role.


Archives of Biochemistry and Biophysics | 2008

Role of αPhe-291 residue in the phosphate-binding subdomain of catalytic sites of Escherichia coli ATP synthase

Laura Brudecki; Johnny J. Grindstaff; Zulfiqar Ahmad

The role of alphaPhe-291 residue in phosphate binding by Escherichia coli F1F0-ATP synthase was examined. X-ray structures of bovine mitochondrial enzyme suggest that this residue resides in close proximity to the conserved betaR246 residue. Herein, we show that mutations alphaF291D and alphaF291E in E. coli reduce the ATPase activity of F1F0 membranes by 350-fold. Yet, significant oxidative phosphorylation activity is retained. In contrast to wild-type, ATPase activities of mutants were not inhibited by MgADP-azide, MgADP-fluoroaluminate, or MgADP-fluoroscandium. Whereas, 7-chloro-4-nitrobenzo-2-oxa-1,3-diazole (NBD-Cl) inhibited wild-type ATPase essentially completely, ATPase in mutants was inhibited maximally by approximately 75%, although reaction still occurred at residue betaTyr-297, proximal to alphaPhe-291 in the phosphate-binding pocket. Inhibition characteristics supported the conclusion that NBD-Cl reacts in betaE (empty) catalytic sites, as shown previously by X-ray structure analysis. Phosphate protected against NBD-Cl inhibition in wild-type but not in mutants. In addition, our data suggest that the interaction of alphaPhe-291 with phosphate during ATP hydrolysis or synthesis may be distinct.


Clinical and Vaccine Immunology | 2013

Mitogen-Activated Protein Kinase Phosphatase 1 Disrupts Proinflammatory Protein Synthesis in Endotoxin-Adapted Monocytes

Laura Brudecki; Donald A. Ferguson; Charles E. McCall; Mohamed El Gazzar

ABSTRACT Autotoxic production of proinflammatory mediators during early sepsis induces excessive inflammation, and their later suppression may limit the immune response. We previously reported that sepsis differentially represses transcription and translation of tumor necrosis factor alpha (TNF-α) and interleukin 1β (IL-1β) to reprogram sepsis inflammation. This switch is gene specific and plays a crucial role in the clinically relevant syndrome of endotoxin adaptation/tolerance, multiorgan failure, and poor sepsis outcome. To further define the mechanisms responsible for translation disruption that follows inflammation induction, we used THP-1 human promonocytes as a model of Toll-like receptor 4 (TLR4) responses found in sepsis. We showed that phosphorylation-dependent activation of p38 mitogen-activated protein kinase (MAPK) and translation disruption of TNF-α and IL-6 follow increased MAPK phosphatase 1 (MKP-1) expression and that MKP-1 knockdown rephosphorylates p38 and restores the capacity to translate TNF-α and IL-6 mRNAs. We also observed that the RNA-binding protein motif 4 (RBM4), a p38 MAPK target, accumulates in an unphosphorylated form in the cytosol in endotoxin-adapted cells, suggesting that dephosphorylated RBM4 may function as a translational repressor. Moreover, MKP-1 knockdown promotes RBM4 phosphorylation, blocks its transfer from the nucleus to the cytosol, and reverses translation repression. We also found that microRNA 146a (miR-146a) knockdown prevents and miR-146a transfection induces MKP-1 expression, which lead to increases or decreases in TNF-α and IL-6 translation, respectively. We conclude that a TLR4-, miR-146a-, p38 MAPK-, and MKP-1-dependent autoregulatory pathway regulates the translation of proinflammatory genes during the acute inflammatory response by spatially and temporally modifying the phosphorylation state of RBM4 translational repressor protein.


Immunology and Cell Biology | 2012

Adoptive transfer of CD34 + cells during murine sepsis rebalances macrophage lipopolysaccharide responses

Laura Brudecki; Donald A. Ferguson; Charles E. McCall; Mohamed El Gazzar

Effective treatment of the acute systemic inflammatory response associated with sepsis is lacking, but likely will require new ways to rebalance dysregulated immune responses. One challenge is that human sepsis often is diagnosed too late to reduce the hyperinflammation of early sepsis. Another is that the sequential response to sepsis inflammation rapidly generates an adaptive and immunosuppressive state, which by epigenetic imprint may last for months or years. Emerging data support that the immunosuppressive phase of sepsis can both directly reprogram gene expression of circulating and tissue cells, and disrupt development and differentiation of myeloid precursor cells into competent immunocytes. We recently reported that adoptive transfer of bone marrow CD34+ cells into mice after sepsis induction by cecal ligation and puncture significantly improves late‐sepsis survival by enhancing bacterial clearance through improved neutrophil and macrophage phagocytosis. That study, however, did not examine whether CD34+ transfer can modify noninfectious acute systemic inflammatory responses. Here, we report that CD34+ cell transfer mice that have survived late sepsis also resist lethal lipopolysaccharide (LPS)‐induced inflammatory shock (88% lived vs 0% of naive mice). The CD34+ cell‐recipient survivor mice administered LPS had globally reduced levels of circulating inflammatory mediators compared with naive mice, but their peritoneal and bone marrow‐derived macrophages (BMDMs), unlike those from naïve mice, remained LPS responsive ex vivo. We further found that CD34+ cell transfer into LPS‐challenged naïve mice had diminished immunosuppression, as assessed by ex vivo responses of peritoneal and BMDMs to LPS challenge. We conclude that CD34+ cell adoptive transfer rebalances dysregulated immune responses associated with sepsis and endotoxin shock.


Journal of Innate Immunity | 2015

Processing Body Formation Limits Proinflammatory Cytokine Synthesis in Endotoxin-Tolerant Monocytes and Murine Septic Macrophages

Clara McClure; Laura Brudecki; Zhi Q. Yao; Charles E. McCall; Mohamed El Gazzar

An anti-inflammatory phenotype with pronounced immunosuppression develops during sepsis, during which time neutrophils and monocytes/macrophages limit their Toll-like receptor 4 responses to bacterial lipopolysaccharide (LPS/endotoxin). We previously reported that during this endotoxin-tolerant state, distinct signaling pathways differentially repress transcription and translation of proinflammatory cytokines such as TNFα and IL-6. Sustained endotoxin tolerance contributes to sepsis mortality. While transcription repression requires chromatin modifications, a translational repressor complex of Argonaute 2 (Ago2) and RNA-binding motif protein 4 (RBM4), which bind the 3′-UTR of TNFα and IL-6 mRNA, limits protein synthesis. Here, we show that Dcp1 supports the assembly of the Ago2 and RBM4 repressor complex into cytoplasmic processing bodies (p-bodies) in endotoxin-tolerant THP-1 human monocytes following stimulation with LPS, resulting in translational repression and limiting protein synthesis. Importantly, this translocation process is reversed by Dcp1 knockdown, which restores TNFα and IL-6 protein levels. We also find this translational repression mechanism in primary macrophages of septic mice. Because p-body formation is a critical step in mRNA translation repression, we conclude that Dcp1 is a major component of the translational repression machinery of endotoxin tolerance and may contribute to sepsis outcome.


The FASEB Journal | 2010

Molecular modulation of the alpha-subunit VISIT-DG sequence in the catalytic sites of Escherichia coli ATP synthase

Zulfiqar Ahmad; Laura Brudecki

Collaboration


Dive into the Laura Brudecki's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mohamed El Gazzar

East Tennessee State University

View shared research outputs
Top Co-Authors

Avatar

Donald A. Ferguson

East Tennessee State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clara McClure

East Tennessee State University

View shared research outputs
Top Co-Authors

Avatar

Alan E. Senior

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Deling Yin

East Tennessee State University

View shared research outputs
Top Co-Authors

Avatar

Gene LeSage

East Tennessee State University

View shared research outputs
Top Co-Authors

Avatar

Johnny J. Grindstaff

East Tennessee State University

View shared research outputs
Top Co-Authors

Avatar

Jonathan P. Moorman

East Tennessee State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge