Uric acid crystals and calcium pyrophosphate dihydrate, the causa

Uric acid crystals and calcium pyrophosphate dihydrate, the causative agents of gout and pseudogout, respectively, were the first crystalline molecules shown to activate the NLRP3 inflammasome

21. Another endogenous molecule, fibrillar amyloid-β, associated with the pathogenesis of Alzheimer’s disease, also activates the NLRP3 Proteasome inhibitor inflammasome in a similar manner 20. Silica and asbestos particles, which cause the fibrotic lung disorders silicosis and asbestosis, respectively, also have been demonstrated to activate the NLRP3 inflammasome 24–26. Additionally, the adjuvant properties of aluminum hydroxide (alum) have been shown to be dependent upon its ability to activate the NLRP3 inflammasome 27–30. The mechanism by which the NLRP3 inflammasome is activated remains unknown. However, two events that are common to all activators of the NLRP3 inflammasome are a potassium efflux and the generation of click here ROS (Fig. 1). Inhibiting the potassium efflux, by increasing extracellular potassium concentrations, results in the abrogation of NLRP3 inflammasome activation 24, 25, 27. The exact role of the potassium efflux is unclear; however, the assembly of the NLRP3 inflammasome may be dependent on a low potassium environment 31. Similarly, inhibition or scavenging

of ROS blocks NLRP3 inflammasome activation (reviewed in 32). Lysosomal membrane disruption following particulate uptake has also been postulated to play a role in NLRP3 inflammasome activation and is reviewed in detail in this issue by Hornung and filipin Latz 33. Necrotic cells release endogenous DAMP that alert the innate immune system to tissue damage. Release of ATP from the necrotic cells is a danger signal that activates the innate immune response. ATP binds the purinergic receptor P2X7 triggering the formation of a pannexin-1 hemichannel, which results in the activation of the NLRP3 inflammasome 34–36. The ability of necrotic cells to activate the NLRP3 inflammasome (Fig. 2) was recently demonstrated

in two independent studies 22, 37. Iyer et al. showed that macrophages challenged with cells that had undergone specific forms of necrotic cell death (pressure-disruption, complement lysis, hypoxic injury) were capable of activating caspase-1 in an NLRP3-dependent manner 22. However, not all methods of necrosis were capable of activating NLRP3; necrotic cells generated by freeze−thaw or UV irradiation failed to activate caspase-1, highlighting the heterogeneity of different mechanisms of necrotic cell death. The ability of NLRP3 to sense cellular damage could also be seen in an in vivo model of renal ischemic acute tubular necrosis 22. Both WT and NLRP3-deficient mice that were subjected to renal ischemia/reperfusion injury displayed similar acute tubular necrosis following injury. However, the subsequent inflammatory response to this necrotic injury was markedly blunted in mice that lacked NLRP3.

Related posts:

  1. Treatment was well tolerated, with no infusion-related or infecti
  2. Monocyte subsets are also critical in complications of atheroscle
  3. Just like their apoptotic counterparts, necrotic cells sometimes externalize pho
  4. Serofendic acid did not regulate rCBF at ischemic and reperfusion
  5. This strategy elicits cycles of mucosal damage, followed by tissu
This entry was posted in Antibody. Bookmark the permalink.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>