Network


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

Hotspot


Dive into the research topics where Eric Suto is active.

Publication


Featured researches published by Eric Suto.


Journal of Clinical Investigation | 2007

In vivo blockade of OX40 ligand inhibits thymic stromal lymphopoietin driven atopic inflammation

Dhaya Seshasayee; Wyne P. Lee; Meijuan Zhou; Jean Shu; Eric Suto; Juan Zhang; Laurie Diehl; Cary D. Austin; Y. Gloria Meng; Martha Tan; Sherron Bullens; Stefan Seeber; Maria E. Fuentes; Aran Frank Labrijn; Yvo Graus; Lisa A. Miller; Edward S. Schelegle; Dallas M. Hyde; Lawren C. Wu; Sarah G. Hymowitz; Flavius Martin

Thymic stromal lymphopoietin (TSLP) potently induces deregulation of Th2 responses, a hallmark feature of allergic inflammatory diseases such as asthma, atopic dermatitis, and allergic rhinitis. However, direct downstream in vivo mediators in the TSLP-induced atopic immune cascade have not been identified. In our current study, we have shown that OX40 ligand (OX40L) is a critical in vivo mediator of TSLP-mediated Th2 responses. Treating mice with OX40L-blocking antibodies substantially inhibited immune responses induced by TSLP in the lung and skin, including Th2 inflammatory cell infiltration, cytokine secretion, and IgE production. OX40L-blocking antibodies also inhibited antigen-driven Th2 inflammation in mouse and nonhuman primate models of asthma. This treatment resulted in both blockade of the OX40-OX40L receptor-ligand interaction and depletion of OX40L-positive cells. The use of a blocking, OX40L-specific mAb thus presents a promising strategy for the treatment of allergic diseases associated with pathologic Th2 immune responses.


Science | 2012

Equilibrative Nucleoside Transporter 3 Deficiency Perturbs Lysosome Function and Macrophage Homeostasis

Chia-Lin Hsu; Wei Yu Lin; Dhaya Seshasayee; Yung-Hsiang Chen; Xiao Ding; Zhonghua Lin; Eric Suto; Zhiyu Huang; Wyne P. Lee; Hyunjoo Park; Min Xu; Mei Sun; Linda Rangell; Jeff Lutman; Sheila Ulufatu; Eric Stefanich; Cecile Chalouni; Meredith Sagolla; Lauri Diehl; Paul J. Fielder; Brian Dean; Mercedesz Balazs; Flavius Martin

From Nucleoside Recycling to Histiocytosis Macrophages remove billions of apoptotic cells daily, releasing their nucleic acid material through lysosomal degradation, which allows the resulting nucleosides to be recycled. Hsu et al. (p. 89, published online 15 December) found that the nucleoside transporter, equilibrative nucleoside transporter 3 (ENT3), was highly expressed in macrophages and showed that mice deficient in this transporter develop histiocytosis and features of lysosomal storage disease. When exposed to apoptotic cells, macrophages carrying human ENT3 mutations accumulated adenosine and increased their lysosomal pH. These changes contributed to an enhanced signaling through macrophage colony-stimulating factor (M-CSF) receptor and, ultimately, to M-CSF–driven myeloproliferative disease. Lack of the transporter critical for recycling of nucleosides after phagocytosis results in a fatal expansion of macrophages. Lysosomal storage diseases (LSDs) are a group of heterogeneous disorders caused by defects in lysosomal enzymes or transporters, resulting in accumulation of undegraded macromolecules or metabolites. Macrophage numbers are expanded in several LSDs, leading to histiocytosis of unknown pathophysiology. Here, we found that mice lacking the equilibrative nucleoside transporter 3 (ENT3) developed a spontaneous and progressive macrophage-dominated histiocytosis. In the absence of ENT3, defective apoptotic cell clearance led to lysosomal nucleoside buildup, elevated intralysosomal pH, and altered macrophage function. The macrophage accumulation was partly due to increased macrophage colony-stimulating factor and receptor expression and signaling secondary to the lysosomal defects. These studies suggest a cellular and molecular basis for the development of histiocytosis in several human syndromes associated with ENT3 mutations and potentially other LSDs.


Clinical Cancer Research | 2013

Development and Preclinical Characterization of a Humanized Antibody Targeting CXCL12

Cuiling Zhong; Jianyong Wang; Bing Li; Hong Xiang; Mark Ultsch; Mary Coons; Terence Wong; Nan Chiang; Suzy Clark; Robyn Clark; Leah Quintana; Peter Gribling; Eric Suto; Kai H. Barck; Racquel Corpuz; Jenny Yao; Rashi Takkar; Wyne P. Lee; Lisa A. Damico-Beyer; Richard D. Carano; Camellia W. Adams; Robert F. Kelley; Weiru Wang; Napoleone Ferrara

Purpose: Our goal was to develop a potent humanized antibody against mouse/human CXCL12. This report summarized its in vitro and in vivo activities. Experimental Design: Cell surface binding and cell migration assays were used to select neutralizing hamster antibodies, followed by testing in several animal models. Monoclonal antibody (mAb) 30D8 was selected for humanization based on its in vitro and in vivo activities. Results: 30D8, a hamster antibody against mouse and human CXCL12α, CXCL12β, and CXCL12γ, was shown to dose-dependently block CXCL12α binding to CXCR4 and CXCR7, and CXCL12α-induced Jurkat cell migration in vitro. Inhibition of primary tumor growth and/or metastasis was observed in several models. 30D8 alone significantly ameliorated arthritis in a mouse collagen-induced arthritis model (CIA). Combination with a TNF-α antagonist was additive. In addition, 30D8 inhibited 50% of laser-induced choroidal neovascularization (CNV) in mice. Humanized 30D8 (hu30D8) showed similar in vitro and in vivo activities as the parental hamster antibody. A crystal structure of the hu30D8 Fab/CXCL12α complex in combination with mutational analysis revealed a “hot spot” around residues Asn44/Asn45 of CXCL12α and part of the RFFESH region required for CXCL12α binding to CXCR4 and CXCR7. Finally, hu30D8 exhibited fast clearance in cynomolgus monkeys but not in rats. Conclusion: CXCL12 is an attractive target for treatment of cancer and inflammation-related diseases; hu30D8 is suitable for testing this hypothesis in humans. Clin Cancer Res; 19(16); 4433–45. ©2013 AACR.


Journal of Immunology | 2015

Conditional Deletion of NF-κB–Inducing Kinase (NIK) in Adult Mice Disrupts Mature B Cell Survival and Activation

Hans Brightbill; Janet Jackman; Eric Suto; Heather S. Kennedy; Charles David Jones; Sreedevi Chalasani; Zhonghua Lin; Lucinda Tam; Meron Roose-Girma; Mercedesz Balazs; Cary D. Austin; Wyne P. Lee; Lawren C. Wu

NF-κB–inducing kinase (NIK) is a primary regulator of the noncanonical NF-κB signaling pathway, which plays a vital role downstream of BAFF, CD40L, lymphotoxin, and other inflammatory mediators. Germline deletion or inactivation of NIK in mice results in the defective development of B cells and secondary lymphoid organs, but the role of NIK in adult animals has not been studied. To address this, we generated mice containing a conditional allele of NIK. Deletion of NIK in adult mice results in decreases in B cell populations in lymph nodes and spleen, similar to what is observed upon blockade of BAFF. Consistent with this, B cells from mice in which NIK is acutely deleted fail to respond to BAFF stimulation in vitro and in vivo. In addition, mice with induced NIK deletion exhibit a significant decrease in germinal center B cells and serum IgA, which is indicative of roles for NIK in additional pathways beyond BAFF signaling. Our conditional NIK-knockout mice may be broadly useful for assessing the postdevelopmental and cell-specific roles of NIK and the noncanonical NF-κB pathway in mice.


Arthritis & Rheumatism | 2015

Dual B Cell Immunotherapy Is Superior to Individual Anti‐CD20 Depletion or BAFF Blockade in Murine Models of Spontaneous or Accelerated Lupus

Wei Yu Lin; Dhaya Seshasayee; Wyne P. Lee; Patrick Caplazi; Sami McVay; Eric Suto; Allen Nguyen; Zhonghua Lin; Yonglian Sun; Laura DeForge; Mercedesz Balazs; Flavius Martin; Ali A. Zarrin

To determine whether a combination of B cell depletion and BAFF blockade is more effective than monotherapy in treating models of spontaneous or accelerated systemic lupus erythematosus (SLE) in (NZB × NZW)F1 mice.


Journal of Leukocyte Biology | 2013

CRIg mediates early Kupffer cell responses to adenovirus

Jeannie Q. He; Kenneth J. Katschke; Peter Gribling; Eric Suto; Wyne P. Lee; Lauri Diehl; Jeffrey Eastham-Anderson; Anusha Ponakala; Laszlo Komuves; Jackson G. Egen; Menno van Lookeren Campagne

Whereas adenoviral vectors are known to activate the complement cascade, leading to fixation of C3 proteins to the viral capsid, the consequences of this activation for viral clearance from the circulation are not known. Liver KCs, the macrophage population responsible for early uptake and elimination of many blood‐borne pathogens, express CRIg, a complement receptor for C3 proteins. Here, we find that CRIg is important for the early elimination of C3‐coated adenoviral vectors from the sinusoidal bloodstream by KCs. We further demonstrate that by acting as a critical receptor for adenovirus phagocytosis, CRIg plays an important role in regulating virus‐induced KC death and depletion of these cells from the liver sinusoidal lumen. Our study thus identifies a critical pathway regulating KC function and survival in response to systemic viral infection.


Journal of Experimental Medicine | 2015

Autoimmunity-associated protein tyrosine phosphatase PEP negatively regulates IFN-α receptor signaling

Derek A. Holmes; Eric Suto; Wyne P. Lee; Qinglin Ou; Qian Gong; Hamish R.C. Smith; Patrick Caplazi; Andrew C. Chan

Holmes et al. demonstrate that the protein tyrosine phosphatase PEP (PTPN22 in humans) associated with lupus and other autoimmune diseases, inhibits IFN-α receptor signaling in mice. PEP-deficient hematopoietic progenitors demonstrate increased IFNAR signaling, IFN-inducible gene expression and proliferation. PEP-deficient mice treated with IFN-α exhibit lupus-like disease and profound defects in hematopoiesis, resulting in cytopenia.


JCI insight | 2017

Btk-specific inhibition blocks pathogenic plasma cell signatures and myeloid cell–associated damage in IFNα-driven lupus nephritis

Arna Katewa; Yugang Wang; Jason A. Hackney; Tao Huang; Eric Suto; Nandhini Ramamoorthi; Cary D. Austin; Meire Bremer; Jacob Chen; James J. Crawford; Kevin S. Currie; Peter A. Blomgren; Jason DeVoss; Julie DiPaolo; Jonathan Hau; Adam R. Johnson; Justin Lesch; Laura DeForge; Zhonghua Lin; Marya Liimatta; Joseph W. Lubach; Sami McVay; Zora Modrusan; Allen Nguyen; Chungkee Poon; Jianyong Wang; Lichuan Liu; Wyne P. Lee; Harvey Wong; Wendy B. Young

Systemic lupus erythematosus (SLE) is often associated with exaggerated B cell activation promoting plasma cell generation, immune-complex deposition in the kidney, renal infiltration of myeloid cells, and glomerular nephritis. Type-I IFNs amplify these autoimmune processes and promote severe disease. Brutons tyrosine kinase (Btk) inhibitors are considered novel therapies for SLE. We describe the characterization of a highly selective reversible Btk inhibitor, G-744. G-744 is efficacious, and superior to blocking BAFF and Syk, in ameliorating severe lupus nephritis in both spontaneous and IFNα-accelerated lupus in NZB/W_F1 mice in therapeutic regimens. Selective Btk inhibition ablated plasmablast generation, reduced autoantibodies, and - similar to cyclophosphamide - improved renal pathology in IFNα-accelerated lupus. Employing global transcriptional profiling of spleen and kidney coupled with cross-species human modular repertoire analyses, we identify similarities in the inflammatory process between mice and humans, and we demonstrate that G-744 reduced gene expression signatures essential for splenic B cell terminal differentiation, particularly the secretory pathway, as well as renal transcriptional profiles coupled with myeloid cell-mediated pathology and glomerular plus tubulointerstitial disease in human glomerulonephritis patients. These findings reveal the mechanism through which a selective Btk inhibitor blocks murine autoimmune kidney disease, highlighting pathway activity that may translate to human SLE.


Journal of Immunology | 2017

The Ox40/Ox40 Ligand Pathway Promotes Pathogenic Th Cell Responses, Plasmablast Accumulation, and Lupus Nephritis in NZB/W F1 Mice

Jonathan Sitrin; Eric Suto; Arthur Wuster; Jeffrey Eastham-Anderson; Jeong M. Kim; Cary D. Austin; Wyne P. Lee; Timothy W. Behrens

Ox40 ligand (Ox40L) locus genetic variants are associated with the risk for systemic lupus erythematosus (SLE); however, it is unclear how Ox40L contributes to SLE pathogenesis. In this study, we evaluated the contribution of Ox40L and its cognate receptor, Ox40, using in vivo agonist and antagonist approaches in the NZB × NZW (NZB/W) F1 mouse model of SLE. Ox40 was highly expressed on several CD4 Th cell subsets in the spleen and kidney of diseased mice, and expression correlated with disease severity. Treatment of aged NZB/W F1 mice with agonist anti-Ox40 mAbs potently exacerbated renal disease, which was accompanied by activation of kidney-infiltrating T cells and cytokine production. The agonist mAbs also induced activation and inflammatory gene expression in splenic CD4 T cells, including IFN-regulated genes, increased the number of follicular helper T cells and plasmablasts in the spleen, and led to elevated levels of serum IgM and enhanced renal glomerular IgM deposition. In a type I IFN–accelerated lupus model, treatment with an antagonist Ox40:Fc fusion protein significantly delayed the onset of severe proteinuria and improved survival. These data support the hypothesis that the Ox40/Ox40L pathway drives cellular and humoral autoimmune responses during lupus nephritis in NZB/W F1 mice and emphasize the potential clinical value of targeting this pathway in human lupus.


Science Signaling | 2015

Inhibition of the kinase ITK in a mouse model of asthma reduces cell death and fails to inhibit the inflammatory response

Yonglian Sun; Ivan Peng; Joshua D. Webster; Eric Suto; Justin Lesch; Xiumin Wu; Kate Senger; George Francis; Kathy Barrett; Jenna L. Collier; Jason D. Burch; Meijuan Zhou; Yuan Chen; Connie Chan; Jeff Eastham-Anderson; Hai Ngu; Olga Li; Tracy Staton; Charles Havnar; Allan Jaochico; Janet Jackman; Surinder Jeet; Lorena Riol-Blanco; Lawren C. Wu; David F. Choy; Joseph R. Arron; Brent S. McKenzie; Nico Ghilardi; Moulay Hicham Alaoui Ismaili; Zhonghua Pei

The kinase ITK is required for antigen-stimulated T cell death. Targeting ITK in asthma CD4+ T helper 2 (TH2) lymphocytes secrete the cytokines interleukin-4 (IL-4), IL-15, and IL-13, which are implicated in the pathogenesis of asthma. Antigen stimulation of T cells activates the kinase ITK, which is required for TH2-type cytokine production. ITK knockout mice are resistant to airway inflammation, which suggests that ITK inhibitors might be used to treat human asthma. However, Sun et al. found that a mouse model of asthma developed worse disease when treated with an ITK-specific inhibitor, exhibiting increased numbers of T cells and amounts of TH2-type cytokines in the airways. These effects were associated with a failure of ITK-inhibited T cells to undergo antigen-stimulated cell death. Together, these data suggest that targeting the kinase activity of ITK in human asthma may exacerbate disease. Interleukin-2 (IL-2)–inducible T cell kinase (ITK) mediates T cell receptor (TCR) signaling primarily to stimulate the production of cytokines, such as IL-4, IL-5, and IL-13, from T helper 2 (TH2) cells. Compared to wild-type mice, ITK knockout mice are resistant to asthma and exhibit reduced lung inflammation and decreased amounts of TH2-type cytokines in the bronchoalveolar lavage fluid. We found that a small-molecule selective inhibitor of ITK blocked TCR-mediated signaling in cultured TH2 cells, including the tyrosine phosphorylation of phospholipase C–γ1 (PLC-γ1) and the secretion of IL-2 and TH2-type cytokines. Unexpectedly, inhibition of the kinase activity of ITK during or after antigen rechallenge in an ovalbumin-induced mouse model of asthma failed to reduce airway hyperresponsiveness and inflammation. Rather, in mice, pharmacological inhibition of ITK resulted in T cell hyperplasia and the increased production of TH2-type cytokines. Thus, our studies predict that inhibition of the kinase activity of ITK may not be therapeutic in patients with asthma.

Collaboration


Dive into the Eric Suto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge