Inflammasomes are multiprotein complexes that are formed and activated after exposure to danger signals with the activation of caspase-1 causing the maturation and release of IL-1β and IL-18. In the formation of inflammasome complexes, several cytosolic pattern recognition receptors (PRRs) act as sensors of pathogen-associated molecular patterns (PAMPS) and damage-associated molecular patterns (DAMPS) [1]. NLRP3-inflammasome is the most described complex consisting of a leucine-rich repeat (LRR) domain, a central nucleotide-binding oligomerization domain (NOD), and an amino-terminal pyrin domain (PYD), which primarily interacts with apoptosis-associated speck-like protein containing a CARD (ASC) [1]. The main interest of the NLRP3-inflammasome is the activity in response to a wide variety of stimuli, such as endogenous danger signals such as extracellular adenosine triphosphate (ATP), uric acid crystals (MSU), and amyloid-β fibrils, free cholesterol, ROS [2]. Many of these stimuli are elevated during ageing, and are implicated in age-related diseases, so the activation of the NLRP3 inflammasome appears to be involved in the production of age-related inflammation. Accordingly, NLRP3-inflammasome has been associated with inflamm-ageing, a low-grade sterile chronic inflammation that has been described as a progressive event during biological ageing with accumulation of pro-inflammatory mediators [3]. Inflammasomes are suspected to play a role in inflammaging, however, their exact contribution to this process remains unknown [4]. Our recent study showed that the genetic deletion of Nlrp3 in mice prolongs lifespan and improves healthspan by attenuating the multiple age-related degenerative changes associated with cardiac and metabolic ageing [5]. Deletion of NLRP3-inflammasome protected aged mice from age-related increase in insulin resistance, reduced IGF-1 and leptin/adiponectin ratio levels. Furthermore, NLRP3 ablation reduced cardiac damage with protection from age-dependent PR interval prolongation, which is associated with atrial fibrillation from cardiovascular ageing and reduced telomere shortening. Interestingly, these KO mice showed inhibition of the PI3K/AKT/mTOR pathway, with consequent improved autophagy flux compared to older wild type mice, and conserved Nampt-mediated NAD+ levels with increased SIRT1 protein expression. So, based on this, the role of the NLRP3-inflammasome during ageing and the inflamm-ageing is very evident. However, the complexity of the inflammasomes shows an independent role of these with respect to the canonical inflammatory pathways. Nlrp3 -/- mice have shown similar levels of other cytokines compared to their littermates controls. Youm et al., showed increased levels of IL-6 in serum from aged Nlrp3 -/- and WT mice [6]. In our recent work, we showed levels similar to serum levels of IL-6, IL-8, TNF, and increased levels of IL-6 protein in cardiac tissues from old mice, but there was a greater increase in heart tissues from old KO mice than WT mice and a similar increase in serum from WT and KO mice (Figure 1A). Interestingly, pharmacological inhibition of Nlrp3 in old mice also shows increased cardiac IL-6 expression (Figure 1B). IL-6, IL-8 and TNF, and their receptors, are upregulated in aged tissues and cells [4]. These findings raise the possibility that the loss of NLRP3 does not affect the age-related increase in other inflammatory pathways and inflamm-ageing, but with the protective effect of the age-related changes that confers the inflammasome an important and independent role in cardiac and metabolic ageing. Therefore, we propose the possibility of an independent aspect of the NLRP3-inflammasome from the inflamm-ageing that could be associated with a new inflammasom-ageing in which the only inhibition of the NLRP3-inflammasome can prevent various aspects related to age.
Figure 1. Changes in the autophagy and inflammation observed in ovaries from young and old mice. (A) Western blot analysis with representative blot of IL-6 levels in cardiac tissue of 3 and 20-mo-old WT and Nlrp3 -/-. (B) Western blot analysis showing the protein expression of inflammatory markers IL-6 in cardiac tissues of old mice after MCC950 treatment. n= 4 mice per group and age. Densitometric analysis is shown as means ± SD. ***P < 0.001, **P < 0.01, young vs old mice.
Further investigation focused on the specific role of NLRP3-inflammasome in cardiac ageing will be required to address these issues and would open a new pharmacological target for the use of the specific NLRP3 inhibitor with respect to anti-inflammatory treatments. We invite to the scientific community to discuss this possible new topic.
References
- 1. Lamkanfi M, Dixit VM. Cell. 2014; 157:1013–22. https://doi.org/10.1016/j.cell.2014.04.007 [PubMed]
- 2. Cordero MD, et al. Trends Endocrinol Metab. 2018; 29:8–17. https://doi.org/10.1016/j.tem.2017.10.009 [PubMed]
- 3. Franceschi C, et al. Ann N Y Acad Sci. 2000; 908:244–54. https://doi.org/10.1111/j.1749-6632.2000.tb06651.x [PubMed]
- 4. Latz E, Duewell P. Semin Immunol. 2018; 40:61–73. https://doi.org/10.1016/j.smim.2018.09.001 [PubMed]
- 5. Marín-Aguilar F, et al. Aging Cell. 2020; 19:e13050. https://doi.org/10.1111/acel.13050 [PubMed]
- 6. Youm YH, et al. Cell Metab. 2013; 18:519–32. https://doi.org/10.1016/j.cmet.2013.09.010 [PubMed]