Thèse de doctorat
Résumé : Type 1 Diabetes (T1D) is an autoimmune disease affecting around 0.1-0.8% of the population worldwide and is characterized by a progressive destruction of insulin-producing beta cells. Pro-inflammatory cytokines released by immune cells around the islets contribute for the “first wave” of beta cell apoptosis. Cytokine-mediated activation of the transcription factor nuclear factor kappa (NF-κB) contributes to beta cell demise in T1D. This is unusual, since NF-κB has anti-apoptotic effects in other cells. NF-κB is activated in most cells via the canonical pathway, while its activation via the non-canonical NF-κB pathway is restricted to few cell types, such as maturing/differentiating immune cell and osteoclasts. We have now observed that IL-1β+IFN-γ induces an atypical activation of the non-canonical NF-κB pathway in beta cells. This activation depends on different crosstalk mechanisms between the canonical and non-canonical NF-κB pathways, including the down-regulation of the E3 ligase Fbw7, which targets the p100 for proteasomal degradation, and up-regulation of another E3 ligase, βTrCP, which in turn induces cleavage of p100 to p52, a hallmark step in the non-canonical NF-κB activation. Importantly, cytokine-mediated activation of the non-canonical pathway regulates the expression of late NF-κB dependent genes, such as Ccl5, Ccl19, Ccl12, Fas that regulate both pro-inflammatory and pro-apoptotic responses and are implicated in beta cell loss in T1D. This atypical activation of the non-canonical NF-κB pathway by pro-inflammatory cytokines in beta cells constitutes a novel “feed-forward” mechanism that may explain the particular pro-apoptotic effect of this transcription factor in beta cells. Besides regulation of pro-death responses, NF-κB activation in beta cells triggers the expression of the ubiquitin-editing protein A20, encoded by TNFAIP3. A20 restricts NF-κB signalling and possess anti-apoptotic activities in beta cells. Importantly, genome-wide association studies have identified TNFAIP3 as a candidate gene for T1D. We presently demonstrated that A20 effects in beta cells are not restricted to inhibition of NF-κB. Thus, A20 also suppresses the pro-apoptotic mitogen-activated protein kinase c-Jun N-terminal kinase (JNK), and activates the survival signaling mediated via the v-akt murine thymoma viral oncogene homolog (Akt), thus inhibiting the intrinsic pathway of apoptosis. Finally, a cohort study of T1D children indicated that the risk allele of the rs2327832 single nucleotide polymorphism of TNFAIP3 predict lower C-peptide and higher hemoglobin A1c (HbA1c) levels 12 months after disease onset, indicating that this candidate gene contributes for reduced residual beta-cell function and impaired glycemic control in early T1D. In conclusion, our results indicate a critical role for A20 in the regulation of beta cell survival and unveil novel mechanisms by which A20 controls beta-cell fate. Moreover, we identified the single nucleotide polymorphism rs2327832 of TNFAIP3 as a prognostic marker for diabetes outcome in children with T1D.We have also observed that A20 protects beta cells against the pro-apoptotic effects of cytokines by preventing the degradation of the anti-apoptotic protein Mcl-1. Mcl-1 belongs to the Bcl-2 family of proteins that regulate the intrinsic apoptotic pathway. It was previously shown that Mcl-1 depletion contributes to apoptosis in rat beta cells and that its expression is downregulated in islets from T1D individuals infected by enteroviruses. We have now confirmed in human beta cells that decreased Mcl-1 expression contributes to cytokine-mediated beta cell death. We generated a beta cell specific Mcl-1 knockout mouse line (βMcl-1 KO) and observed that islets derived from these mice were more susceptible to pro-apoptotic stimuli exposure ex vivo. Of note, βMcl-1 KO mice were more vulnerable to multiple low dose streptozotocin-induced diabetes than their wild type littermates. One of the mechanisms by which cytokines mediate Mcl-1 degradation is via its phosphorylation by GSK3β. Overexpression of A20 increased AKT phosphorylation, providing a negative feedback on GSK3β activity and preventing Mcl-1 degradation. Cytokines also increase Mcl-1 ubiquitination, a process regulated by the E3 ligases Mule and βTrCP and the deubiquitinase USP9X. The present findings indicate that pro-inflammatory cytokines trigger post-translational modifications of Mcl-1 leading to its degradation. This contributes to exacerbation of pro-death responses and beta cell demise in T1D, but it can be prevented, at least in part, by A20. As a whole, our data unveil novel post-translational mechanisms and different ubiquitin editing proteins that regulate beta cell fate in T1D and are modulated by pro-inflammatory cytokines.