Cell death is an integral part of the immune responses to pathogenic infection, and different forms of cell death can have different immunological consequences. For example, apoptosis is thought to clear viral infected cells and to control run-away immune responses whereas pyroptosis of macrophages can result in the release of cytokines. The Tumor Necrosis Factor Receptor (TNF-R) family of death receptors can trigger apoptosis but also an alternative form of death called programmed necrosis or necroptosis. Necroptosis is dependent on the RIP1 and RIP3 kinases but little is known on its physiological role. To address this, we have generated three lines of tissue- specific FADD knockout mice. FADD is a known adapter protein for all the TNF-R death receptors and thus FADD-deficient cells are resistant to death receptor-mediated apoptosis. However, stimulation of FADD-deficient cells leads to death through necroptosis. This occurs in T-cell receptor stimulated T cells from T-cell specific-FADD knockout (tFADD-/-) mice, in lipopolysaccharide (LPS) stimulated dendritic cells (DCs) from DC-specific-FADD deficient (dcFADD-/-) mice and in LPS stimulated macrophages from macrophage-specific FADD deficient (mFADD-/-) mice. T cells from tFADD-/- mice are functionally defective due to premature necroptosis. However, FADD-deficient DCs are functionally normal and can secrete cytokines when stimulated. Analysis of dcFADD-/- mice showed that some of their phenotypes are surprisingly similar to DC-less mice with imbalance erythrocytes and myeloproliferative disease. At the same time, dcFADD-/- mice exhibit a modest increase of inflammation. In contrast to other DC-specific apoptosis-resistant mice, aged dcFADD-/- mice don't suffer from autoimmunity but they appear to have an enhanced immune system. We hypothesize that necroptosis is one of the strategies for innate immune cells to stimulate the immune system in cases when receptor-induced apoptosis is blocked. In dcFADD-/- mice, necroptotic DCs releasing inflammatory contents can lead to improved immunity against pathogenic infection. This hypothesis will be tested in aim 1. Using DC-specific FADD-/-/MyD88-/- and dcFADD-/-/RIP3-/- mice, we will address whether constitutive stimulation of DCs through TLRs leads to necroptosis and release of inflammatory contents. To examine the possible enhanced immunity of dcFADD-/- mice, they will be challenged with influenza and Toxoplasma gondii and studied in details. Whether necroptosis is required for immunity against these two pathogens will be assessed using RIP3-/- mice.
In Aim 2, we will unravel the biochemical signaling pathways involving FADD in necroptosis in response to TLR stimulation. Successful completion of these aims will greatly enhance our understanding of the host- pathogen interaction and the physiological role of necroptosis.

Public Health Relevance

Cell death, including apoptosis, pyroptosis, autophagic cell death and necrosis, is an integral part of development of the mouse/human immune system and/or responses to pathogenic infection. Members of the tumor necrosis factor (TNF) death receptors can initiate apoptosis and a new form of necrosis called necroptosis but the physiological role of necroptosis is poorly understood; we propose in this application to understand the role of necroptosis in infection by studying mouse models deficient in an adapter protein required for the TNF cell death signals.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
4R01AI095299-05
Application #
8997965
Study Section
Cellular and Molecular Immunology - B Study Section (CMIB)
Program Officer
Singleton, Kentner L
Project Start
2012-03-02
Project End
2017-02-28
Budget Start
2016-03-01
Budget End
2017-02-28
Support Year
5
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of California Berkeley
Department
Internal Medicine/Medicine
Type
Schools of Arts and Sciences
DUNS #
124726725
City
Berkeley
State
CA
Country
United States
Zip Code
94704
Schock, Suruchi N; Chandra, Neha V; Sun, Yuefang et al. (2017) Induction of necroptotic cell death by viral activation of the RIG-I or STING pathway. Cell Death Differ 24:615-625
Schock, Suruchi N; Young, Jennifer A; He, Tina H et al. (2015) Deletion of FADD in macrophages and granulocytes results in RIP3- and MyD88-dependent systemic inflammation. PLoS One 10:e0124391
Young, Jennifer A; He, Tina H; Reizis, Boris et al. (2013) Commensal microbiota are required for systemic inflammation triggered by necrotic dendritic cells. Cell Rep 3:1932-44