2010
DOI: 10.1038/nature09413
|View full text |Cite
|
Sign up to set email alerts
|

Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis

Abstract: Apoptotic cells release ‘find-me’ signals at the earliest stages of death to recruit phagocytes1. The nucleotides ATP and UTP represent one class of find-me signals2, but their mechanism of release is not known. Here, we identify the plasma membrane channel pannexin 1 (PANX1) as a mediator of find-me signal/nucleotide release from apoptotic cells. Pharmacological inhibition and siRNA-mediated knockdown of PANX1 led to decreased nucleotide release and monocyte recruitment by apoptotic cells. Conversely, PANX1 o… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1

Citation Types

40
1,142
5
7

Year Published

2013
2013
2024
2024

Publication Types

Select...
6
1

Relationship

1
6

Authors

Journals

citations
Cited by 930 publications
(1,201 citation statements)
references
References 33 publications
40
1,142
5
7
Order By: Relevance
“…45 The administration of cumate to tPANX1-transfected, but not to control, MCA205 cells rendered them permeable to the small fluorescent dye YO-PRO-1, but not to the vital dye DAPI (Figure 3c), in line with the previously reported capacity of tPANX1 to promote a selective permeabilization of the plasma membrane. [32][33][34] The expression of tPANX1 drove a YO-PRO-1 influx coupled to an ATP efflux that could be inhibited in a dose-dependent manner by monensin and DIDS but not by Y-27632, blebbistatin and Z-VAD-fmk (Figures 3d and e), confirming the specificity of this panel of inhibitors.…”
Section: Panx1 Channels Operate Independently From Autophagysupporting
confidence: 57%
See 3 more Smart Citations
“…45 The administration of cumate to tPANX1-transfected, but not to control, MCA205 cells rendered them permeable to the small fluorescent dye YO-PRO-1, but not to the vital dye DAPI (Figure 3c), in line with the previously reported capacity of tPANX1 to promote a selective permeabilization of the plasma membrane. [32][33][34] The expression of tPANX1 drove a YO-PRO-1 influx coupled to an ATP efflux that could be inhibited in a dose-dependent manner by monensin and DIDS but not by Y-27632, blebbistatin and Z-VAD-fmk (Figures 3d and e), confirming the specificity of this panel of inhibitors.…”
Section: Panx1 Channels Operate Independently From Autophagysupporting
confidence: 57%
“…[32][33][34] Surprisingly, however, the Figure 7 Lysosomes and autophago(lyso)somes participate in the trafficking and/or in the maintenance of the intracellular ATP pool. (a-c) Human osteosarcoma U2OS cells were transfected with a non-targeting siRNA (siUNR) or with the indicated siRNAs for 48 h, and then maintained in control conditions (Co) or treated with 4 mM mitoxantrone (MTX) or 300 mM oxaliplatin (OXA) for additional 18 h. Finally, cells were processed for the fluorescence microscopy-assisted visualization of nuclei (with Hoechst 33342, blue fluorescence), ATP-containing vesicles (with quinacrine, green fluorescence) or LAMP1 (revealed with an secondary antibody emitting in red) (a and b), and ATP levels in culture supernatants assessed by a luciferase-based test (c).…”
Section: Discussionmentioning
confidence: 99%
See 2 more Smart Citations
“…The mechanism involved incorporates the release of ATP and UTP molecules into the extracellular space, leading to the activation of P2X7 receptors (Virginio et al 1999). The opening of cation channels follows, allowing entry of YP1, as well as other large molecules (Chekeni et al 2010;Michel et al 2000;Virginio et al 1999). Functional activity of P2X7 receptors can influence apoptotic pathways (Chow et al 1997), and activation of these receptors has been used to promote cell death in cancer cells (Gorodeski 2009).…”
Section: Introductionmentioning
confidence: 99%