Network


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

Hotspot


Dive into the research topics where Marat Khodoun is active.

Publication


Featured researches published by Marat Khodoun.


The Journal of Allergy and Clinical Immunology | 2011

Ingested allergens must be absorbed systemically to induce systemic anaphylaxis

Richard T. Strait; Ashley Mahler; Simon P. Hogan; Marat Khodoun; Akira Shibuya; Fred D. Finkelman

BACKGROUND IgE-mediated food allergy is a common cause of enteric disease and is responsible for approximately 100 systemic anaphylaxis deaths in the United States each year. IgG antibodies can protect against IgE-mediated systemic anaphylaxis induced by injected antigens by neutralizing antigens before they can bind to mast cell-associated IgE. OBJECTIVE We have investigated whether IgA and IgG antibodies can similarly protect against systemic, IgE-mediated anaphylaxis induced by ingested antigens and, if so, whether IgA and IgG antibodies protect by neutralizing antigens before or after their systemic absorption. METHODS Murine passive and active anaphylaxis models were used to study the abilities of serum versus gut lumenal IgA antibodies and serum IgG antibodies to inhibit systemic anaphylaxis induced by ingested allergens in normal mice, mice deficient in the ability to secrete IgA into the intestines, and mice in which intestinal IL-9 overexpression has induced intestinal mastocytosis and increased intestinal permeability. RESULTS IgE-mediated systemic anaphylaxis and mast cell degranulation induced by antigen ingestion are suppressed by both serum antigen-specific IgA and IgG, but not by IgA within the gut lumen. CONCLUSION Systemic rather than enteric antibodies protect against systemic anaphylaxis induced by ingested antigen. This implies that ingested antigens must be absorbed systemically to induce anaphylaxis and suggests that immunization protocols that increase serum levels of antigen-specific, non-IgE antibodies should protect against severe food allergy.


European Journal of Immunology | 2015

Differential control of CD4+ T-cell subsets by the PD-1/PD-L1 axis in a mouse model of allergic asthma

Jaclyn W. McAlees; Stephane Lajoie; Krista Dienger; Alyssa Sproles; Phoebe K. Richgels; Yanfen Yang; Marat Khodoun; Miyuki Azuma; Hideo Yagita; Patricia C. Fulkerson; Marsha Wills-Karp; Ian P. Lewkowich

Studies examining the role of PD‐1 family members in allergic asthma have yielded conflicting results. Using a mouse model of allergic asthma, we demonstrate that blockade of PD‐1/PD‐L1 has distinct influences on different CD4+ T‐cell subsets. PD‐1/PD‐L1 blockade enhances airway hyperreactivity (AHR), not by altering the magnitude of the underlying Th2‐type immune response, but by allowing the development of a concomitant Th17‐type immune response. Supporting differential CD4+ T‐cell responsiveness to PD‐1‐mediated inhibition, naïve PD‐1−/− mice displayed elevated Th1 and Th17 levels, but diminished Th2 cytokine levels, and ligation of PD‐1 in WT cells limited cytokine production by in vitro polarized Th1 and Th17 cells, but slightly enhanced cytokine production by in vitro polarized Th2 cells. Furthermore, PD‐1 ligation enhanced Th2 cytokine production by naïve T cells cultured under nonpolarizing conditions. These data demonstrate that different CD4+ T‐cell subsets respond differentially to PD‐1 ligation and may explain some of the variable results observed in control of allergic asthma by the PD‐1 family members. As the PD‐1/PD‐L1 axis limits asthma severity by constraining Th17 cell activity, this suggests that severe allergic asthma may be associated with a defective PD‐1/PD‐L1 regulatory axis in some individuals.


The Journal of Allergy and Clinical Immunology | 2013

Rapid desensitization of mice with anti-FcγRIIb/FcγRIII mAb safely prevents IgG-mediated anaphylaxis

Marat Khodoun; Zeynep Yesim Kucuk; Richard T. Strait; Durga Krishnamurthy; Kevin Janek; Corey D. Clay; Suzanne C. Morris; Fred D. Finkelman

BACKGROUND Stimulatory IgG receptors (FcγRs) on bone marrow-derived cells contribute to the pathogenesis of several autoimmune and inflammatory disorders. Monoclonal antibodies that block FcγRs might suppress these diseases, but they can induce anaphylaxis. OBJECTIVE We wanted to determine whether a rapid desensitization approach can safely suppress IgG/FcγR-mediated anaphylaxis. METHODS Mice were injected with serially increasing doses of 2.4G2, a rat mAb that blocks the inhibitory FcγR, FcγRIIb, and the stimulatory receptor, FcγRIII. Rectal temperature was used to detect the development of anaphylaxis. Passive and active IgG-mediated anaphylaxis were evaluated in mice that had been rapidly desensitized with 2.4G2 or mock-desensitized in mice in which monocyte/macrophages, basophils, or neutrophils had been depleted or desensitized and in mice in which FcγRI, FcγRIII, and/or FcγRIV had been deleted or blocked. RESULTS Rapid desensitization with 2.4G2 prevented 2.4G2-induced shock and completely suppressed IgG-mediated anaphylaxis. Rapid desensitization of ovalbumin-sensitized mice with 2.4G2 was safer and more effective than rapid desensitization with ovalbumin. 2.4G2 treatment completely blocked FcγRIII and removed most FcγRI and FcγRIV from nucleated peripheral blood cells. Because IgG(2a)-mediated anaphylaxis was partially FcγRI and FcγRIV dependent, the effects of 2.4G2 on FcγRI and FcγRIV were probably crucial for its complete inhibition of IgG(2a)-mediated anaphylaxis. IgG(2a)-mediated anaphylaxis was partially inhibited by depletion or desensitization of monocyte/macrophages, basophils, or neutrophils. CONCLUSION IgG-mediated anaphylaxis can be induced by ligation of FcγRI, FcγRIII, or FcγRIV on monocycte/macrophages, basophils, or neutrophils and can be safely suppressed by rapid desensitization with anti-FcγRII/RIII mAb. A similar approach may safely suppress other FcγR-dependent immunopathology.


The Journal of Allergy and Clinical Immunology | 2013

Rapid polyclonal desensitization with antibodies to IgE and FcεRIα.

Marat Khodoun; Zeynep Yesim Kucuk; Richard T. Strait; Durga Krishnamurthy; Kevin Janek; Ian P. Lewkowich; Suzanne C. Morris; Fred D. Finkelman

BACKGROUND Rapid desensitization, a procedure in which persons allergic to an antigen are treated at short intervals with increasing doses of that antigen until they tolerate a large dose, is an effective, but risky, way to induce temporary tolerance. OBJECTIVE We wanted to determine whether this approach can be adapted to suppress all IgE-mediated allergies in mice by injecting serially increasing doses of monoclonal antibodies (mAbs) to IgE or FcεRIα. METHODS Active and passive models of antigen- and anti-IgE mAb-induced IgE-mediated anaphylaxis were used. Mice were desensitized with serially increasing doses of anti-IgE mAb, anti-FcεRIα mAb, or antigen. Development of shock (hypothermia), histamine and mast cell protease release, cytokine secretion, calcium flux, and changes in cell number and FcεRI and IgE expression were evaluated. RESULTS Rapid desensitization with anti-IgE mAb suppressed IgE-mediated immediate hypersensitivity; however, some mice developed mild anaphylaxis during desensitization. Rapid desensitization with anti-FcεRIα mAb that only binds FcεRI that is not occupied by IgE suppressed both active and passive IgE-mediated anaphylaxis without inducing disease. It quickly, but temporarily, suppressed IgE-mediated anaphylaxis by decreasing mast cell signaling through FcεRI, then slowly induced longer lasting mast cell unresponsiveness by removing membrane FcεRI. Rapid desensitization with anti-FcεRIα mAb was safer and longer lasting than rapid desensitization with antigen. CONCLUSION A rapid desensitization approach with anti-FcεRIα mAb safely desensitizes mice to IgE-mediated anaphylaxis by inducing mast cell anergy and later removing all mast cell IgE. Rapid desensitization with an anti-human FcεRIα mAb may be able to prevent human IgE-mediated anaphylaxis.


Toxicology | 2014

Glyphosate–rich air samples induce IL–33, TSLP and generate IL–13 dependent airway inflammation

Sudhir Kumar; Marat Khodoun; Eric M. Kettleson; Christopher McKnight; Tiina Reponen; Sergey A. Grinshpun; Atin Adhikari

Several low weight molecules have often been implicated in the induction of occupational asthma. Glyphosate, a small molecule herbicide, is widely used in the world. There is a controversy regarding a role of glyphosate in developing asthma and rhinitis among farmers, the mechanism of which is unexplored. The aim of this study was to explore the mechanisms of glyphosate induced pulmonary pathology by utilizing murine models and real environmental samples. C57BL/6, TLR4-/-, and IL-13-/- mice inhaled extracts of glyphosate-rich air samples collected on farms during spraying of herbicides or inhaled different doses of glyphosate and ovalbumin. The cellular response, humoral response, and lung function of exposed mice were evaluated. Exposure to glyphosate-rich air samples as well as glyphosate alone to the lungs increased: eosinophil and neutrophil counts, mast cell degranulation, and production of IL-33, TSLP, IL-13, and IL-5. In contrast, in vivo systemic IL-4 production was not increased. Co-administration of ovalbumin with glyphosate did not substantially change the inflammatory immune response. However, IL-13-deficiency resulted in diminished inflammatory response but did not have a significant effect on airway resistance upon methacholine challenge after 7 or 21 days of glyphosate exposure. Glyphosate-rich farm air samples as well as glyphosate alone were found to induce pulmonary IL-13-dependent inflammation and promote Th2 type cytokines, but not IL-4 for glyphosate alone. This study, for the first time, provides evidence for the mechanism of glyphosate-induced occupational lung disease.


The Journal of Allergy and Clinical Immunology | 2017

Anti-FcγRIIB mAb suppresses murine IgG-dependent anaphylaxis by Fc domain targeting of FcγRIII

Corey D. Clay; Richard T. Strait; Ashley Mahler; Marat Khodoun; Fred D. Finkelman

Background: The inhibitory receptor Fc&ggr;RIIB is expressed on human and murine bone marrow–derived cells and limits inflammation by suppressing signaling through stimulatory receptors. Objective: We sought to evaluate the effects of K9.361, a mouse IgG2a alloantibody to mouse Fc&ggr;RIIB, on murine anaphylaxis. Methods: Wild‐type and Fc&ggr;R‐deficient mice were used to study anaphylaxis, which was induced by injection of 2.4G2 (rat IgG2b mAb that binds both Fc&ggr;RIIB and the stimulatory receptor Fc&ggr;RIII), by actively immunizing IgE‐deficient mice and then challenging with the immunizing antigen, and by passive immunization with IgG or IgE anti–2,4,6‐trinitrophenyl mAb, followed by injection of 2,4,6‐trinitrophenyl–ovalbumin. Pretreatment with K9.361 was assessed for its ability to influence anaphylaxis. Results: Unexpectedly, K9.361 injection induced mild anaphylaxis, which was both Fc&ggr;RIIB and Fc&ggr;RIII dependent and greatly enhanced by &bgr;‐adrenergic blockade. K9.361 injection also decreased expression of stimulatory Fc&ggr; receptors, especially Fc&ggr;RIII, and strongly suppressed IgG‐mediated anaphylaxis without strongly affecting IgE‐mediated anaphylaxis. The F(ab′)2 fragment of K9.361 did not induce anaphylaxis, even after &bgr;‐adrenergic blockade, and did not deplete Fc&ggr;RIII or suppress IgG‐mediated anaphylaxis but prevented intact K9.361‐induced anaphylaxis without diminishing intact K9.36 suppression of IgG‐mediated anaphylaxis. Conclusion: Cross‐linking Fc&ggr;RIIB to stimulatory Fc&ggr;Rs through the Fc domains of an anti‐Fc&ggr;RIIB mAb induces and then suppresses IgG‐mediated anaphylaxis without affecting IgE‐mediated anaphylaxis. Because IgG‐ and IgE‐mediated anaphylaxis can be mediated by the same cell types, this suggests that desensitization acts at the receptor rather than cellular level. Sequential treatment with the F(ab′)2 fragment of anti‐Fc&ggr;RIIB mAb followed by intact anti‐Fc&ggr;RIIB safely prevents IgG‐mediated anaphylaxis.


Archive | 2012

Methods for suppressing allergic reactions

Fred D. Finkelman; Marat Khodoun; Zeynep Yesim Kucuk; Durga Krishnmurthy; Richard T. Strait


Archive | 2008

Methods of modulating inflammation and compositions therefore

Fred D. Finkelman; Marat Khodoun; Marsha Willis-Karp; Christina Lewis


Journal of Immunology | 2016

Suppression of established food allergy by a combination of anti-TSLP, anti-IL-33, and anti-IL-25 monoclonal antibodies.

Marat Khodoun; Durga Krishnamurthy; Richard T. Strait; Joel Tocker; Crystal Potter; Fred D. Finkelman


Archive | 2015

Methods and Compositions for Suppressing IgE-Mediated Anaphylaxis

Fred D. Finkelman; Marat Khodoun; Suzanne C. Morris

Collaboration


Dive into the Marat Khodoun's collaboration.

Top Co-Authors

Avatar

Fred D. Finkelman

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard T. Strait

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zeynep Yesim Kucuk

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ashley Mahler

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Corey D. Clay

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Ian P. Lewkowich

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kevin Janek

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Simon P. Hogan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge