Network


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

Hotspot


Dive into the research topics where Gregorio Gomez is active.

Publication


Featured researches published by Gregorio Gomez.


Journal of Experimental Medicine | 2004

Negative Regulation of Immunoglobulin E–dependent Allergic Responses by Lyn Kinase

Sandra Odom; Gregorio Gomez; Martina Kovarova; Yasuko Furumoto; John J. Ryan; Harry V. Wright; Claudia Gonzalez-Espinosa; Margaret L. Hibbs; Kenneth W. Harder; Juan Rivera

A role for Lyn kinase as a positive regulator of immunoglobulin (Ig)E-dependent allergy has long been accepted. Contrary to this belief, Lyn kinase was found to have an important role as a negative regulator of the allergic response. This became apparent from the hyperresponsive degranulation of lyn − / − bone marrow–derived mast cells, which is driven by hyperactivation of Fyn kinase that occurs, in part, through the loss of negative regulation by COOH-terminal Src kinase (Csk) and the adaptor, Csk-binding protein. This phenotype is recapitulated in vivo as young lyn − / − mice showed an enhanced anaphylactic response. In vivo studies also demonstrated that as lyn − / − mice aged, their serum IgE increased as well as occupancy of the high affinity IgE receptor (FcεRI). This was mirrored by increased circulating histamine, increased mast cell numbers, increased cell surface expression of the high affinity IgE receptor (FcεRI), and eosinophilia. The increased IgE production was not a consequence of increased Fyn kinase activity in lyn − / − mice because both lyn − / − and lyn − / − fyn − / − mice showed high IgE levels. Thus, lyn − / − mice and mast cells thereof show multiple allergy-associated traits, causing reconsideration of the possible efficacy in therapeutic targeting of Lyn in allergic disease.


Journal of Immunology | 2005

Impaired FcRI-Dependent Gene Expression and Defective Eicosanoid and Cytokine Production as a Consequence of Fyn Deficiency in Mast Cells 1

Gregorio Gomez; Claudia Gonzalez-Espinosa; Sandra Odom; Gabriela Baez; M. Eugenia Cid; John J. Ryan; Juan Rivera

Fyn kinase is a key contributor in coupling FcεRI to mast cell degranulation. A limited macroarray analysis of FcεRI-induced gene expression suggested potential defects in lipid metabolism, eicosanoid and glutathione metabolism, and cytokine production. Biochemical analysis of these responses revealed that Fyn-deficient mast cells failed to secrete the inflammatory eicosanoid products leukotrienes B4 and C4, the cytokines IL-6 and TNF, and chemokines CCL2 (MCP-1) and CCL4 (MIP-1β). FcεRI-induced generation of arachidonic acid and normal induction of cytokine mRNA were defective. Defects in JNK and p38 MAPK activation were observed, whereas ERK1/2 and cytosolic phospholipase A2 (S505) phosphorylation was normal. Pharmacological studies revealed that JNK activity was associated with generation of arachidonic acid. FcεRI-mediated activation of IκB kinase β and IκBα phosphorylation and degradation was defective resulting in a marked decrease of the nuclear NF-κB DNA binding activity that drives IL-6 and TNF production in mast cells. However, not all cytokine were affected, as IL-13 production and secretion was enhanced. These studies reveal a major positive role for Fyn kinase in multiple mast cell inflammatory responses and demonstrate a selective negative regulatory role for certain cytokines.


Journal of Experimental Medicine | 2003

The Four Distal Tyrosines Are Required for LAT-dependent Signaling in FcεRI-mediated Mast Cell Activation

Shin-ichiroh Saitoh; Sandra Odom; Gregorio Gomez; Connie L. Sommers; Howard A. Young; Juan Rivera; Lawrence E. Samelson

The linker for activation of T cells (LAT) is an adaptor protein critical for FcɛRI-mediated mast cell activation. LAT is a substrate of the tyrosine kinases activated after TCR and FcɛRI engagement. After phosphorylation of the cytosolic domain of LAT, multiple signaling molecules such as phospholipase C–γ1, Grb2, and Gads associate with phosphorylated LAT via their SH2 domains. The essential role of the four distal tyrosines in TCR-mediated signaling and T cell development has been demonstrated by experiments using LAT-deficient cell lines and genetically modified mice. To investigate the role of these four tyrosines of LAT in FcɛRI-mediated mast cell activation, bone marrow–derived mast cells from LAT-deficient mice were infected with retroviral vectors designed to express wild-type or mutant LAT. Examination of bone marrow–derived mast cells expressing various tyrosine to phenylalanine mutants in LAT demonstrates a differential requirement for these different binding sites. In these studies, assays of biochemical pathways, degranulation, and cytokine and chemokine release were performed. Finally, the role of these tyrosines was also evaluated in vivo using genetically modified animals. Deletion of all four distal tyrosines, and in particular, loss of the primary phospholipase C–γ-binding tyrosine had a significant effect on antigen-induced histamine release.


Immunity | 2009

Lyn Kinase Controls Basophil GATA-3 Transcription Factor Expression and Induction of Th2 Cell Differentiation

Nicolas Charles; Wendy T. Watford; Haydeé L. Ramos; Lars Hellman; Hans C. Oettgen; Gregorio Gomez; John J. Ryan; John J. O'Shea; Juan Rivera

T helper 1 (Th1)-Th2 cell balance is key to host defense and its dysregulation has pathophysiological consequences. Basophils are important in Th2 cell differentiation. However, the factors controlling the onset and extent of basophil-mediated Th2 cell differentiation are unknown. Here, we demonstrate that Lyn kinase dampened basophil expression of the transcription factor GATA-3 and the initiation and extent of Th2 cell differentiation. Lyn-deficient mice had a marked basophilia, a constitutive Th2 cell skewing that was exacerbated upon in vivo challenge of basophils, produced antibodies to a normally inert antigen, and failed to appropriately respond to a Th1 cell-inducing pathogen. The Th2 cell skewing was dependent on basophils, immunoglobulin E, and interleukin-4, but was independent of mast cells. Our findings demonstrate that basophil-expressed Lyn kinase exerts regulatory control on Th2 cell differentiation and function.


Journal of Immunology | 2005

TGF-β1 Inhibits Mast Cell FcεRI Expression

Gregorio Gomez; Carlos D. Ramirez; Juan Rivera; Manish Patel; Farnaz Norozian; Harry V. Wright; Mohit Kashyap; Brian Barnstein; Krista Fischer-Stenger; Lawrence B. Schwartz; Christopher L. Kepley; John J. Ryan

Mast cell activation through the high affinity IgE receptor (FcεRI) is a critical component of atopic inflammation. The cytokine TGF-β1 has been shown to inhibit IgE-dependent mast cell activation, possibly serving to dampen mast cell-mediated inflammatory responses. We present proof that TGF-β1 inhibits mast cell FcεRI expression through a reversible pathway that diminishes protein, but not mRNA, expression of the FcεRI subunit proteins α, β, and γ. The stability of the expressed proteins and the assembled cell surface complex was unaltered by TGF-β1 treatment. However, TGF-β1 decreased the rate of FcεRI β-chain synthesis, arguing that this inhibitory cytokine exerts its effects at the level of mRNA translation. TGF-β1 consistently diminished FcεRI expression on cultured human or mouse mast cells as well as freshly isolated peritoneal mast cells. The related cytokines, TGF-β2 and TGF-β3, had similar effects. We propose that TGF-β1 acts as a negative regulator of mast cell function, in part by decreasing FcεRI expression.


Journal of Immunology | 2004

IL-10 Inhibits FcεRI Expression in Mouse Mast Cells

Sheila R. Gillespie; Randall R. DeMartino; Jingfang Zhu; Hey Jin Chong; Carlos D. Ramirez; Christopher P. Shelburne; L.Andrew Bouton; Daniel P. Bailey; Anita Gharse; Paria Mirmonsef; Sandra Odom; Gregorio Gomez; Juan Rivera; Krista Fischer-Stenger; John J. Ryan

FcεRI expression and function is a central aspect of allergic disease. Using bone marrow-derived mouse mast cell populations, we have previously shown that the Th2 cytokine IL-4 inhibits FcεRI expression and function. In the current study we show that the Th2 cytokine IL-10 has similar regulatory properties, and that it augments the inhibitory effects of IL-4. FcεRI down-regulation was functionally significant, as it diminished inflammatory cytokine production and IgE-mediated FcεRI up-regulation. IL-10 and IL-4 reduced FcεRI β protein expression without altering the α or γ subunits. The ability of IL-4 and IL-10 to alter FcεRI expression by targeting the β-chain, a critical receptor subunit known to modulate receptor expression and signaling, suggests the presence of a Th2 cytokine-mediated homeostatic network that could serve to both initiate and limit mast cell effector function.


Journal of Immunology | 2007

Omalizumab Reverses the Phenotypic and Functional Effects of IgE-Enhanced FcεRI on Human Skin Mast Cells

Gregorio Gomez; Sherryline Jogie-Brahim; Mika Shima; Lawrence B. Schwartz

The dramatic effects of the anti-IgE mAb omalizumab to lower free IgE levels and FcεRI levels on basophils contrast with more modest clinical effects. Accordingly, whether IgE modulates FcεRI levels and FcεRI-dependent mediator release in vitro on human skin mast cells (MCTC type) that had matured in vivo is of interest. IgE reversibly enhanced FcεRI levels on MCTC cells in a dose- and time-dependent manner (up-regulation t1/2 of 4–5 days with 1–3 μg/ml IgE), without affecting cell proliferation. A molar ratio of omalizumab to IgE of 0.9 at baseline prevented receptor up-regulation by 50%, whereas adding omalizumab to MCTC cells already with IgE-enhanced FcεRI levels at molar ratios of 5, 12.5, and 31 reduced FcεRI levels to baseline with respective t1/2 values of 8.7, 6.3, and 4.8 days. MCTC cells with IgE-enhanced FcεRI levels were more sensitive to stimulation with a low dose of anti-FcεRI mAb in terms of degranulation and production of PGD2, GM-CSF, IL-6, IL-13, and TNF-α. Reducing up-regulated FcεRI levels with omalizumab also reduced mediator release to a low dose of anti-FcεRI mAb to baseline by 3–4 wk. Thus, reducing free IgE should decrease the hypersensitivity of allergic individuals to low naturally occurring concentrations of allergens.


Journal of Immunology | 2008

TGF-β1 Attenuates Mediator Release and De Novo Kit Expression by Human Skin Mast Cells through a Smad-Dependent Pathway

Wei Zhao; Gregorio Gomez; Shao-Hua Yu; John J. Ryan; Lawrence B. Schwartz

TGF-β has pleiotropic effects on many cell types at different stages of their development, including mast cells. The present study examines the effects of TGF-β on human skin mast cells of the MCTC type. The expression of TGF-β receptors (TGF-R) was verified at the mRNA and protein levels for TGF-RI and TGF-RII, and at the mRNA level for accessory molecules β-glycan and endoglin. TGF-β did not affect mast cell viability after 1 wk at concentrations ≤10 ng/ml, but at 50 ng/ml caused significant cell death. TGF-β inhibited surface and total expression of Kit in a dose-dependent manner, whereas the surface expression of FcεRI, FcγRI, and FcγRII was not affected. TGF-β inhibited degranulation and cytokine production, but not PGD2 production. TGF-β diminished surface Kit expression through a TGF-RI kinase/Smad-dependent pathway by inhibiting new synthesis of Kit protein, which became evident following internalization and degradation of Kit after mast cells were exposed to the Kit ligand, stem cell factor. In contrast, addition of TGF-β had no discernible effect on surface Kit expression when administered 3 days after stem cell factor, by which time surface Kit levels had returned to baseline. Although both transcription and translation are important for de novo expression of Kit, Kit mRNA levels were not affected by TGF-β. Therefore, transcription of a gene other than Kit might be involved in Kit expression. Finally, activation of mast cells increased their susceptibility to TGF-β-mediated apoptosis, a process that might regulate the survival of activated mast cells in vivo.


Journal of Immunology | 2010

IL-4 and TGF-β1 Counterbalance One Another while Regulating Mast Cell Homeostasis

Matthew Macey; Jamie Sturgill; Johanna K. Morales; Yves T. Falanga; Joshua Morales; Sarah K. Norton; Nitin Yerram; Hoon Shim; Josephine Fernando; Alasdair M. Gifillan; Gregorio Gomez; Lawrence B. Schwartz; Carole A. Oskeritzian; Sarah Spiegel; Daniel H. Conrad; John J. Ryan

Mast cell responses can be altered by cytokines, including those secreted by Th2 and regulatory T cells (Treg). Given the important role of mast cells in Th2-mediated inflammation and recent demonstrations of Treg-mast cell interactions, we examined the ability of IL-4 and TGF-β1 to regulate mast cell homeostasis. Using in vitro and in vivo studies of mouse and human mast cells, we demonstrate that IL-4 suppresses TGF-β1 receptor expression and signaling, and vice versa. In vitro studies demonstrated that IL-4 and TGF-β1 had balancing effects on mast cell survival, migration, and FcεRI expression, with each cytokine cancelling the effects of the other. However, in vivo analysis of peritoneal inflammation during Nippostrongylus brasiliensis infection in mice revealed a dominant suppressive function for TGF-β1. These data support the existence of a cytokine network involving the Th2 cytokine IL-4 and the Treg cytokine TGF-β1 that can regulate mast cell homeostasis. Dysregulation of this balance may impact allergic disease and be amenable to targeted therapy.


Journal of Clinical Immunology | 2011

Disparity in FcεRI-Induced Degranulation of Primary Human Lung and Skin Mast Cells Exposed to Adenosine

Gregorio Gomez; Wei Zhao; Lawrence B. Schwartz

Inhaled and intravenously administered adenosine induces mast cell-mediated (histamine-dependent) bronchospasm in asthmatics without causing urticaria. A differential response to adenosine by human lung and skin mast cells is shown: low concentrations potentiate FcεRI-induced degranulation of human lung mast cells but not that of skin mast cells. Human lung mast cells were found to express ∼3-fold more A3AR messenger RNA (mRNA) than skin mast cells, suggesting the involvement of the Gi-linked A3AR. Indeed, the adenosine-induced potentiation was sensitive to inhibition by pertussis toxin and, furthermore, could be induced with an A3AR-specific agonist. This study reveals a previously unrecognized disparity in the response to adenosine by primary human mast cells from lung and skin that might explain why adenosine induces a pulmonary but not dermatologic allergy-like response in vivo. In addition, we identify the A3AR as a potentiating receptor of FcεRI-induced degranulation, thereby implicating it in the in vivo bronchoconstrictive response to adenosine in asthmatics.

Collaboration


Dive into the Gregorio Gomez's collaboration.

Top Co-Authors

Avatar

Lawrence B. Schwartz

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

John J. Ryan

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Juan Rivera

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sandra Odom

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wei Zhao

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel P. Bailey

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Martina Kovarova

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Mohit Kashyap

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge