Cargando…

NLRP12 is an innate immune checkpoint for repressing IFN signatures and attenuating lupus nephritis progression

Signaling driven by nucleic acid sensors participates in interferonopathy-mediated autoimmune diseases. NLRP12, a pyrin-containing NLR protein, is a negative regulator of innate immune activation and type I interferon (IFN-I) production. Peripheral blood mononuclear cells (PBMCs) derived from system...

Descripción completa

Detalles Bibliográficos
Autores principales: Tsao, Yen-Po, Tseng, Fang-Yu, Chao, Chih-Wei, Chen, Ming-Han, Yeh, Yi-Chen, Abdulkareem, Babamale Olarewaju, Chen, Se-Yi, Chuang, Wen-Ting, Chang, Pei-Ching, Chen, I-Chun, Wang, Pin-Hsuan, Wu, Chien-Sheng, Tsai, Chang-Youh, Chen, Szu-Ting
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Clinical Investigation 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9888378/
https://www.ncbi.nlm.nih.gov/pubmed/36719379
http://dx.doi.org/10.1172/JCI157272
Descripción
Sumario:Signaling driven by nucleic acid sensors participates in interferonopathy-mediated autoimmune diseases. NLRP12, a pyrin-containing NLR protein, is a negative regulator of innate immune activation and type I interferon (IFN-I) production. Peripheral blood mononuclear cells (PBMCs) derived from systemic lupus erythematosus (SLE) patients expressed lower levels of NLRP12, with an inverse correlation with IFNA expression and high disease activity. NLRP12 expression was transcriptionally suppressed by runt-related transcription factor 1–dependent (RUNX1-dependent) epigenetic regulation under IFN-I treatment, which enhanced a negative feedback loop between low NLRP12 expression and IFN-I production. Reduced NLRP12 protein levels in SLE monocytes was linked to spontaneous activation of innate immune signaling and hyperresponsiveness to nucleic acid stimulations. Pristane-treated Nlrp12(–/–) mice exhibited augmented inflammation and immune responses; and substantial lymphoid hypertrophy was characterized in NLRP12-deficient lupus-prone mice. NLRP12 deficiency mediated the increase of autoantibody production, intensive glomerular IgG deposition, monocyte recruitment, and the deterioration of kidney function. These were bound in an IFN-I signature–dependent manner in the mouse models. Collectively, we reveal a remarkable link between low NLRP12 expression and lupus progression, which suggests the impact of NLRP12 on homeostasis and immune resilience.