Network


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

Hotspot


Dive into the research topics where Ivan Peng is active.

Publication


Featured researches published by Ivan Peng.


Nature | 2014

A Crohn/'s disease variant in Atg16l1 enhances its degradation by caspase 3

Aditya Murthy; Yun Li; Ivan Peng; Mike Reichelt; Anand K. Katakam; Rajkumar Noubade; Merone Roose-Girma; Jason DeVoss; Lauri Diehl; Robert R. Graham; Menno van Lookeren Campagne

Crohn’s disease is a debilitating inflammatory bowel disease (IBD) that can involve the entire digestive tract. A single-nucleotide polymorphism (SNP) encoding a missense variant in the autophagy gene ATG16L1 (rs2241880, Thr300Ala) is strongly associated with the incidence of Crohn’s disease. Numerous studies have demonstrated the effect of ATG16L1 deletion or deficiency; however, the molecular consequences of the Thr300Ala (T300A) variant remains unknown. Here we show that amino acids 296–299 constitute a caspase cleavage motif in ATG16L1 and that the T300A variant (T316A in mice) significantly increases ATG16L1 sensitization to caspase-3-mediated processing. We observed that death-receptor activation or starvation-induced metabolic stress in human and murine macrophages increased degradation of the T300A or T316A variants of ATG16L1, respectively, resulting in diminished autophagy. Knock-in mice harbouring the T316A variant showed defective clearance of the ileal pathogen Yersinia enterocolitica and an elevated inflammatory cytokine response. In turn, deletion of the caspase-3-encoding gene, Casp3, or elimination of the caspase cleavage site by site-directed mutagenesis rescued starvation-induced autophagy and pathogen clearance, respectively. These findings demonstrate that caspase 3 activation in the presence of a common risk allele leads to accelerated degradation of ATG16L1, placing cellular stress, apoptotic stimuli and impaired autophagy in a unified pathway that predisposes to Crohn’s disease.


Nature Medicine | 2009

Targeted depletion of lymphotoxin-alpha-expressing TH1 and TH17 cells inhibits autoimmune disease.

Eugene Y. Chiang; Ganesh Kolumam; Xin Yu; Michelle Francesco; Sinisa Ivelja; Ivan Peng; Peter Gribling; Jean Shu; Wyne P. Lee; Canio J. Refino; Mercedesz Balazs; Andres Paler-Martinez; Allen Nguyen; Judy Young; Kai H. Barck; Richard A. D. Carano; Ron Ferrando; Lauri Diehl; Devavani Chatterjea; Jane L. Grogan

Uncontrolled T helper type 1 (TH1) and TH17 cells are associated with autoimmune responses. We identify surface lymphotoxin-α (LT-α) as common to TH0, TH1 and TH17 cells and employ a unique strategy to target these subsets using a depleting monoclonal antibody (mAb) directed to surface LT-α. Depleting LT-α–specific mAb inhibited T cell–mediated models of delayed-type hypersensitivity and experimental autoimmune encephalomyelitis. In collagen-induced arthritis (CIA), preventive and therapeutic administration of LT-α–specific mAb inhibited disease, and immunoablated T cells expressing interleukin-17 (IL-17), interferon-γ and tumor necrosis factor-α (TNF-α), whereas decoy lymphotoxin-β receptor (LT-βR) fusion protein had no effect. A mutation in the Fc tail, rendering the antibody incapable of Fcγ receptor binding and antibody-dependent cellular cytotoxicity activity, abolished all in vivo effects. Efficacy in CIA was preceded by a loss of rheumatoid-associated cytokines IL-6, IL-1β and TNF-α within joints. These data indicate that depleting LT-α–expressing lymphocytes with LT-α–specific mAb may be beneficial in the treatment of autoimmune disease.


Journal of Immunology | 2010

IL-17RC Is Required for IL-17A– and IL-17F–Dependent Signaling and the Pathogenesis of Experimental Autoimmune Encephalomyelitis

Yan Hu; Naruhisa Ota; Ivan Peng; Canio J. Refino; Dimitry M. Danilenko; Patrick Caplazi; Wenjun Ouyang

It has been suggested that IL-17RC forms a complex with IL-17RA to mediate the functions of IL-17A and IL-17F homodimers as well as IL-17A/F heterodimers. It is still unclear whether IL-17RC is absolutely required for the signaling of IL-17 cytokines in vivo. By using Il-17rc–deficient mice, we show that IL-17RC is essential for the signaling of IL-17A, IL-17F, and IL-17A/F both in vitro and in vivo. IL-17RC does not preassociate with IL-17RA on the cell surface; rather IL-17A can induce the formation of an IL-17RC and IL-17RA complex. This process is not dependent on the intracellular similar expression to fibroblast growth factor genes and IL-17Rs (SEFIR) domain of IL-17RC, but the SEFIR is essential in IL-17A signal transduction. Finally, Il-17rc−/− mice develop much milder disease in an experimental autoimmune encephalomyelitis model, supporting an essential role for IL-17RC in mediating immune-mediated CNS inflammation.


Nature | 2014

NRROS negatively regulates reactive oxygen species during host defence and autoimmunity.

Rajkumar Noubade; Kit Wong; Naruhisa Ota; Sascha Rutz; Céline Eidenschenk; Patricia Valdez; Jiabing Ding; Ivan Peng; Andrew Sebrell; Patrick Caplazi; Jason DeVoss; Robert Soriano; Tao Sai; Rongze Lu; Zora Modrusan; Jason A. Hackney; Wenjun Ouyang

Reactive oxygen species (ROS) produced by phagocytes are essential for host defence against bacterial and fungal infections. Individuals with defective ROS production machinery develop chronic granulomatous disease. Conversely, excessive ROS can cause collateral tissue damage during inflammatory processes and therefore needs to be tightly regulated. Here we describe a protein, we termed negative regulator of ROS (NRROS), which limits ROS generation by phagocytes during inflammatory responses. NRROS expression in phagocytes can be repressed by inflammatory signals. NRROS-deficient phagocytes produce increased ROS upon inflammatory challenges, and mice lacking NRROS in their phagocytes show enhanced bactericidal activity against Escherichia coli and Listeria monocytogenes. Conversely, these mice develop severe experimental autoimmune encephalomyelitis owing to oxidative tissue damage in the central nervous system. Mechanistically, NRROS is localized to the endoplasmic reticulum, where it directly interacts with nascent NOX2 (also known as gp91phox and encoded by Cybb) monomer, one of the membrane-bound subunits of the NADPH oxidase complex, and facilitates the degradation of NOX2 through the endoplasmic-reticulum-associated degradation pathway. Thus, NRROS provides a hitherto undefined mechanism for regulating ROS prodution—one that enables phagocytes to produce higher amounts of ROS, if required to control invading pathogens, while minimizing unwanted collateral tissue damage.


Journal of Immunology | 2010

Actin-Bundling Protein L-Plastin Regulates T Cell Activation

Chen Wang; Sharon Celeste Morley; David L. Donermeyer; Ivan Peng; Wyne P. Lee; Jason DeVoss; Dimitry M. Danilenko; Zhonghua Lin; Juan Zhang; Jie Zhou; Paul M. Allen; Eric J. Brown

Engagement of TCRs induces actin rearrangements, which are critical for T cell activation. T cell responses require new actin polymerization, but the significance of higher-order actin structures, such as microfilament bundles, is unknown. To determine the role of the actin-bundling protein leukocyte-plastin (L-plastin; LPL) in this process, T cells from LPL−/− mice were studied. LPL−/− T cells were markedly defective in TCR-mediated cytokine production and proliferation. LPL−/− T cells also spread inefficiently on surfaces with immobilized TCR ligands and formed smaller immunological synapses with APCs, likely due to defective formation of lamellipodia. LPL−/− mice showed delayed rejection of skin allografts after release from immunosuppression. Moreover, LPL−/− mice developed much less severe neurologic symptoms in experimental autoimmune encephalomyelitis, which correlated with impaired T cell responses to Ag, manifested by reduced proliferation and production of IFN-γ and IL-17. Thus, LPL-dependent actin bundling facilitates the formation of lamellipodia and normal immunological synapses and thereby enables T cell activation.


Journal of Medicinal Chemistry | 2013

Lead Optimization of a 4-Aminopyridine Benzamide Scaffold To Identify Potent, Selective, and Orally Bioavailable TYK2 Inhibitors.

Jun Liang; A van Abbema; Mercedesz Balazs; Kathy Barrett; L Berezhkovsky; Wade S. Blair; Christine Chang; Donnie Delarosa; Jason DeVoss; J Driscoll; Charles Eigenbrot; Nico Ghilardi; Paul Gibbons; Jason S. Halladay; Amber E. Johnson; Pawan Bir Kohli; Yingjie Lai; Y Liu; Joseph P. Lyssikatos; Priscilla Mantik; Kapil Menghrajani; Jeremy Murray; Ivan Peng; Amy Sambrone; Steven Shia; Young G. Shin; Jan Smith; Sue Sohn; Tsui; Mark Ultsch

Herein we report our lead optimization effort to identify potent, selective, and orally bioavailable TYK2 inhibitors, starting with lead molecule 3. We used structure-based design to discover 2,6-dichloro-4-cyanophenyl and (1R,2R)-2-fluorocyclopropylamide modifications, each of which exhibited improved TYK2 potency and JAK1 and JAK2 selectivity relative to 3. Further optimization eventually led to compound 37 that showed good TYK2 enzyme and interleukin-12 (IL-12) cell potency, as well as acceptable cellular JAK1 and JAK2 selectivity and excellent oral exposure in mice. When tested in a mouse IL-12 PK/PD model, compound 37 showed statistically significant knockdown of cytokine interferon-γ (IFNγ), suggesting that selective inhibition of TYK2 kinase activity might be sufficient to block the IL-12 pathway in vivo.


Molecular Pharmaceutics | 2015

An Anti-B7-H4 Antibody–Drug Conjugate for the Treatment of Breast Cancer

Steven R. Leong; Wei Ching Liang; Yan Wu; Lisa Crocker; Eric Cheng; Deepak Sampath; Rachana Ohri; Helga Raab; Philip E. Hass; Thinh Pham; Ron Firestein; Dongwei Li; Melissa Schutten; Nicola J. Stagg; Annie Ogasawara; Neelima Koppada; Leslie Roth; Simon Williams; Byoung Chul Lee; Cecile Chalouni; Ivan Peng; Jason DeVoss; Jarrod Tremayne; Paul Polakis; Andrew G. Polson

B7-H4 has been implicated in cancers of the female reproductive system and investigated for its possible use as a biomarker for cancer, but there are no preclinical studies to demonstrate that B7-H4 is a molecular target for therapeutic intervention of cancer. We provide evidence that the prevalence and expression levels of B7-H4 are high in different subtypes of breast cancer and that only a few normal tissues express B7-H4 on the cell membrane. These profiles of low normal expression and upregulation in cancer provide an opportunity for the use of antibody-drug conjugates (ADCs), cytotoxic drugs chemically linked to antibodies, for the treatment of B7-H4 positive cancers. We have developed an ADC specific to B7-H4 that uses a linker drug consisting of a potent antimitotic, monomethyl auristatin E (MMAE), linked to engineered cysteines (THIOMAB) via a protease labile linker. We will refer to ADCs that use the THIOMAB format as TDCs to help distinguish the format from standard MC-vc-MMAE ADCs that are conjugated to the interchain disulfide bonds. Anti-B7-H4 (h1D11)-MC-vc-PAB-MMAE (h1D11 TDC) produced durable tumor regression in cell line and patient-derived xenograft models of triple-negative breast cancer. It also binds rat B7-H4 with similar affinity to human and allowed us to test for target dependent toxicity in rats. We found that our anti-B7-H4 TDC has toxicity findings similar to untargeted TDC. Our results validate B7-H4 as an ADC target for breast cancer and support the possible use of this TDC in the treatment of B7-H4(+) breast cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Dendritic cells require NIK for CD40-dependent cross-priming of CD8+ T cells.

Anand K. Katakam; Hans Brightbill; Christian Franci; Chung Kung; Victor Nunez; Charles David Jones; Ivan Peng; Surinder Jeet; Lawren C. Wu; Ira Mellman; Lélia Delamarre; Cary D. Austin

Significance The noncanonical NF-κB signaling pathway via the serine kinase NIK (NF-κB–inducing kinase) is essential for normal immune system development and has been implicated in tumor cell survival and growth. Because NIK is under investigation as a therapeutic target, it is important to understand NIK’s role in the context of a fully developed immune system, particularly in regard to mounting adaptive T-cell responses. We have generated and characterized transgenic mice with conditionally deleted NIK in CD11c+ dendritic cells and observe impaired antigen cross-priming of a naive CD8 T-cell response. This defect results from defective antigen cross-presentation by CD8+ dendritic cells and also is associated with their reduced ability to secrete IL-12p40, a cytokine known to promote cross-priming in vivo. Dendritic cells (DCs) link innate and adaptive immunity and use a host of innate immune and inflammatory receptors to respond to pathogens and inflammatory stimuli. Although DC maturation via canonical NF-κB signaling is critical for many of these functions, the role of noncanonical NF-κB signaling via the serine/threonine kinase NIK (NF-κB–inducing kinase) remains unclear. Because NIK-deficient mice lack secondary lymphoid organs, we generated transgenic mice with targeted NIK deletion in CD11c+ cells. Although these mice exhibited normal lymphoid organs, they were defective in cross-priming naive CD8+ T cells following vaccination, even in the presence of anti-CD40 or polyinosinic:polycytidylic acid to induce DC maturation. This impairment reflected two intrinsic defects observed in splenic CD8+ DCs in vitro, namely antigen cross-presentation to CD8+ T cells and secretion of IL-12p40, a cytokine known to promote cross-priming in vivo. In contrast, antigen presentation to CD4+ T cells was not affected. These findings reveal that NIK, and thus probably the noncanonical NF-κB pathway, is critical to allow DCs to acquire the capacity to cross-present antigen and prime CD8 T cells after exposure to licensing stimuli, such as an agonistic anti-CD40 antibody or Toll-like receptor 3 ligand.


Journal of Immunology | 2014

PILRα Negatively Regulates Mouse Inflammatory Arthritis

Yonglian Sun; Patrick Caplazi; Juan Zhang; Anita Mazloom; Sarah K. Kummerfeld; Gabriel Quinones; Kate Senger; Justin Lesch; Ivan Peng; Andrew Sebrell; Wilman Luk; Yanmei Lu; Zhonghua Lin; Kai H. Barck; Judy Young; Mariela Del Rio; Sophie M. Lehar; Vida Asghari; Wei Yu Lin; Sanjeev Mariathasan; Jason DeVoss; Shahram Misaghi; Mercedesz Balazs; Tao Sai; Benjamin Haley; Philip E. Hass; Min Xu; Wenjun Ouyang; Flavius Martin; Wyne P. Lee

Paired Ig-like type 2 receptor (PILR)α inhibitory receptor and its counterpart PILRβ activating receptor are coexpressed on myeloid cells. In this article, we report that PILRα, but not PILRβ, is elevated in human rheumatoid arthritis synovial tissue and correlates with inflammatory cell infiltration. Pilrα−/− mice produce more pathogenic cytokines during inflammation and are prone to enhanced autoimmune arthritis. Correspondingly, engaging PILRα with anti-PILRα mAb ameliorates inflammation in mouse arthritis models and suppresses the production of proinflammatory cytokines. Our studies suggest that PILRα mediates an important inhibitory pathway that can dampen inflammatory responses.


Nature Communications | 2017

Potassium channels Kv1.3 and KCa3.1 cooperatively and compensatorily regulate antigen-specific memory T cell functions

Eugene Y. Chiang; Tianbo Li; Surinder Jeet; Ivan Peng; Juan Zhang; Wyne P. Lee; Jason DeVoss; Patrick Caplazi; Jun Chen; Søren Warming; David H. Hackos; Susmith Mukund; Christopher M. Koth; Jane L. Grogan

Voltage-gated Kv1.3 and Ca2+-dependent KCa3.1 are the most prevalent K+ channels expressed by human and rat T cells. Despite the preferential upregulation of Kv1.3 over KCa3.1 on autoantigen-experienced effector memory T cells, whether Kv1.3 is required for their induction and function is unclear. Here we show, using Kv1.3-deficient rats, that Kv1.3 is involved in the development of chronically activated antigen-specific T cells. Several immune responses are normal in Kv1.3 knockout (KO) rats, suggesting that KCa3.1 can compensate for the absence of Kv1.3 under these specific settings. However, experiments with Kv1.3 KO rats and Kv1.3 siRNA knockdown or channel-specific inhibition of human T cells show that maximal T-cell responses against autoantigen or repeated tetanus toxoid stimulations require both Kv1.3 and KCa3.1. Finally, our data also suggest that T-cell dependency on Kv1.3 or KCa3.1 might be irreversibly modulated by antigen exposure.

Collaboration


Dive into the Ivan Peng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Caplazi

Washington State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric J. Brown

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge