Overall, my laboratory investigates the interactions of chemokine mimics with G-protein coupled chemotactic receptors and activating receptors with resultant effects on inflammation, immunity, autoimmunity, cancer and algesia. We have shown that a variety of antimicrobial peptides (AMPs) and nuclear binding proteins that mimic chemokines also have the capacity to rapidly activate host immune responses. We have proposed calling these early warning signals alarmins. Alarmins are characterized by having chemotactic activity for cells expressing GiPCR, together with the capacity to interact with another receptor resulting in the activation of immature dendritic cells (iDC) to mature into antigen- presenting, T lymphocyte activating dendritic cells (mDC) with resultant in vivo immunoadjuvant effects. These alarmins, if administered together with an antigen, result in considerable augmentation of both cellular and humoral in vivo immune responses to the antigen. We previously identified both alpha and beta types of defensins as alarmins with chemotactic and activating effects on immature dendritic cells (iDC) and in vivo immunoadjuvant effects. Some of the beta defensins interact with the CCR6 chemokine receptor, others with CCR2, while alpha defensins interact with an as yet unknown G-Protein Coupled Receptors (GiPCR). Another antimicrobial peptide known as cathelicidin (LL37) and its murine homologue CRAMP are chemotactic for FPR2 receptors expressed on monocytes and precursors of iDC, induce the maturation of iDC and are equally as potent adjuvants in vivo as alum. In addition, we have previously also identified eosinophil derived neurotoxin (EDN, a ribonuclease), granulysin from lymphocytes, lactoferrin from neutrophils and HMGB1, a nuclear binding protein as functional alarmins. Although alarmins are structurally distinct, they are rapidly released from granules of leukocytes and epithelial cells or from damaged cells. Alarmins can also be induced in response to proinflammatory stimulants by keratinocytes or epithelial cells lining the GI tract, GU tract and tracheobronchial tree. As such, alarmins probably represent an early warning system to alert the host defense to danger signals During the past three years, we have identified and characterized High Mobility Group Nucleasome-binding protein-1 (HMGN-1) as an extracellular alarmin that is a necessary mediator of lipopolysaccharide (LPS)-induced (TLR-4 dependent) immune responses. HMGN-1 has the capacity to recruit and induce the maturation of dendritic cells (DC) at sites of injection. HMGN-1 activates NF kappa B and multiple MAP kinases largely in a TLR4 dependent manner. Upon coadministration with antigens, HMGN-1 markedly enhances specific immune responses and has potent adjuvant effects favoring Th1 immune responses. Conversely, mice engineered to be deficient in HMGN-1 had greatly reduced antigen specific immune responses even in response to antigens administered together with LPS. This immune deficiency of HMGN-1 knockout mice was associated with deficient recruitment of DC to sites of immunization and reduced cytokine production by DC. Thus, HMGN-1 which is largely derived from non-leukocytes (e.g. epithelial cells) plays a non-redundant critical role in the development of innate and adaptive immune responses. Consequently, HMGN-1 knockout mice also have been shown to spontaneously develop tumors more frequently than normal and to exhibit reduced resistance to tumor challenge. We therefore plan to utilize HMGN-1 as an antitumor vaccine adjuvant.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC009369-21
Application #
8348933
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
21
Fiscal Year
2011
Total Cost
$1,072,297
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Arts, Rob J W; Huang, Po-Kai; Yang, De et al. (2018) High-Mobility Group Nucleosome-Binding Protein 1 as Endogenous Ligand Induces Innate Immune Tolerance in a TLR4-Sirtuin-1 Dependent Manner in Human Blood Peripheral Mononuclear Cells. Front Immunol 9:526
Yang, De; Han, Zhen; Alam, Md Masud et al. (2018) High-mobility group nucleosome binding domain 1 (HMGN1) functions as a Th1-polarizing alarmin. Semin Immunol :
Stolzenberg, Ethan; Berry, Deborah; Yang, De et al. (2017) A Role for Neuronal Alpha-Synuclein in Gastrointestinal Immunity. J Innate Immun 9:456-463
Yang, De; Han, Zhen; Oppenheim, Joost J (2017) Alarmins and immunity. Immunol Rev 280:41-56
Nie, Yingjie; Yang, De; Trivett, Anna et al. (2017) Development of a Curative Therapeutic Vaccine (TheraVac) for the Treatment of Large Established Tumors. Sci Rep 7:14186
Yoon, Juhan; Leyva-Castillo, Juan Manuel; Wang, Guoxing et al. (2016) IL-23 induced in keratinocytes by endogenous TLR4 ligands polarizes dendritic cells to drive IL-22 responses to skin immunization. J Exp Med 213:2147-66
Nie, Yingjie; Yang, De; Oppenheim, Joost J (2016) Alarmins and Antitumor Immunity. Clin Ther 38:1042-53
Wang, Fang; Qiao, Linan; Lv, Xing et al. (2016) Alarmin human ? defensin HNP1 activates plasmacytoid dendritic cells by triggering NF-?B and IRF1 signaling pathways. Cytokine 83:53-60
Chen, Xin; Willette-Brown, Jami; Wu, Xueqiang et al. (2015) IKK? is required for the homeostasis of regulatory T cells and for the expansion of both regulatory and effector CD4 T cells. FASEB J 29:443-54
Oppenheim, Joost J (2014) Evolution of the serendipitous discovery of macrophage-lymphocyte interactions. Front Immunol 5:530

Showing the most recent 10 out of 32 publications