Return to search

The Role of c-FLIP in the Regulation of Apoptosis, Necroptosis and Autophagy in T Lymphocytes

<p>To maintain homeostasis, T lymphocytes die through caspase&ndash;dependent apoptosis. However, blockage of caspase activity in T lymphocytes does not increase cell survival. The loss of caspase 8 activity leads to programmed necrosis (necroptosis) upon T cell receptor (TCR) stimulation in T lymphocytes. Necroptosis is correlated with excessive macroautophagy, an intracellular catabolic process characterized by the sequestration of cytoplasmic compartments through double&ndash;membrane vacuoles. Meanwhile, the proper induction of macroautophagy is required for T lymphocyte survival and function. Cellular caspase 8 (FLICE)&ndash;like inhibitory protein (c&ndash;FLIP) promotes survival in T lymphocytes. c&ndash;FLIP suppresses death receptor&ndash;induced apoptosis by modulating caspase 8 activation. Whether this modulation plays a role in the regulation of necroptosis has yet to be studied. Additionally, overexpression of c&ndash;FLIP reduces autophagy induction and promotes cell survival in cell lines. It remains unclear whether c&ndash;FLIP protects primary T lymphocytes by regulating the threshold at which autophagy occurs. In this study, c&ndash;FLIP isoform&ndash;specific conditional deletion models were used to study the role of c&ndash;FLIP in necroptosis and autophagy in primary T lymphocytes.</p><p>Our results showed that the long isoform of c&ndash;FLIP (c&ndash; FLIP<sub>L</sub>) regulates necroptosis by inhibiting receptor interacting protein 1 (RIP&ndash;1). Upon TCR stimulation, c&ndash;FLIP<sub>L</sub>&ndash;deficient T cells underwent RIP&ndash;1&ndash;dependent necroptosis. Interestingly, though previous studies have generally described necroptosis in the absence of caspase 8 activity and apoptosis, pro&ndash;apoptotic caspase 8 activity and the rate of apoptosis were also increased in c&ndash;FLIPL&ndash;deficient T lymphocytes. Moreover, c&ndash; FLIP<sub>L</sub>&ndash;deficient T cells exhibited enhanced autophagy, which served a cytoprotective function. </p><p>Apoptosis can be induced by either death receptors on the plasma membrane (extrinsic pathway), or the damage of the genome and/or cellular organelles (intrinsic pathway). Previous studies in c&ndash;FLIP&ndash;deficient T lymphocytes suggested that c&ndash;FLIP promotes cell survival in the absence of death receptor signals. Independent of death receptor signaling, mitochondria sense apoptotic stimuli and mediate the activation of caspases. Whether c&ndash;FLIP regulates mitochondrion&ndash;dependent apoptotic signaling remains unknown. Here, by deleting the <italic>c&ndash;Flip <italic> gene in mature T lymphocytes, we showed a role for c&ndash;FLIP in the intrinsic apoptosis pathway. In naïve T cells stimulated with the apoptosis inducer, c&ndash;FLIP suppressed cytochrome c release from mitochondria. Bim&ndash;deletion rescued the enhanced apoptosis in c&ndash;FLIP&ndash;deficient T cells, while inhibition of caspase 8 did not. Different from activated T cells, there were no signs of necroptosis in c&ndash;FLIP&ndash;deficient naïve T cells. Together, our findings indicate that c&ndash;FLIP is a key regulator of apoptosis, necroptosis and autophagy in T lymphocytes.</p> / Dissertation

Identiferoai:union.ndltd.org:DUKE/oai:dukespace.lib.duke.edu:10161/8222
Date January 2013
CreatorsHe, Ming-Xiao
ContributorsHe, You-Wen
Source SetsDuke University
Detected LanguageEnglish
TypeDissertation

Page generated in 0.0021 seconds