Back to Search Start Over

Altered responsiveness to extracellular ATP enhances acetaminophen hepatotoxicity

Authors :
Jayane L. D. Quintão
Frederico Marianetti Soriani
Mauro M. Teixeira
Cristina Bonorino
Cristiano Xavier Lima
Rafael Fernandes Zanin
Daniele Araújo Pires
Sylvia S. Amaral
Juliana Gil Melgaço
Maria de Fátima Leite
Bruna R. Sousa
Gustavo B. Menezes
André G. Oliveira
Pedro Marques
Marcelo Alves Pinto
Denise Carmona Cara
Rodrigo R. Resende
Lídia M. Andrade
Ariane C Gomes
Rafaela Vaz Sousa Pereira
Remo Castro Russo
Source :
Cell Communication and Signaling : CCS
Publication Year :
2013
Publisher :
BMC, 2013.

Abstract

Background Adenosine triphosphate (ATP) is secreted from hepatocytes under physiological conditions and plays an important role in liver biology through the activation of P2 receptors. Conversely, higher extracellular ATP concentrations, as observed during necrosis, trigger inflammatory responses that contribute to the progression of liver injury. Impaired calcium (Ca2+) homeostasis is a hallmark of acetaminophen (APAP)-induced hepatotoxicity, and since ATP induces mobilization of the intracellular Ca2+ stocks, we evaluated if the release of ATP during APAP-induced necrosis could directly contribute to hepatocyte death. Results APAP overdose resulted in liver necrosis, massive neutrophil infiltration and large non-perfused areas, as well as remote lung inflammation. In the liver, these effects were significantly abrogated after ATP metabolism by apyrase or P2X receptors blockage, but none of the treatments prevented remote lung inflammation, suggesting a confined local contribution of purinergic signaling into liver environment. In vitro, APAP administration to primary mouse hepatocytes and also HepG2 cells caused cell death in a dose-dependent manner. Interestingly, exposure of HepG2 cells to APAP elicited significant release of ATP to the supernatant in levels that were high enough to promote direct cytotoxicity to healthy primary hepatocytes or HepG2 cells. In agreement to our in vivo results, apyrase treatment or blockage of P2 receptors reduced APAP cytotoxicity. Likewise, ATP exposure caused significant higher intracellular Ca2+ signal in APAP-treated primary hepatocytes, which was reproduced in HepG2 cells. Quantitative real time PCR showed that APAP-challenged HepG2 cells expressed higher levels of several purinergic receptors, which may explain the hypersensitivity to extracellular ATP. This phenotype was confirmed in humans analyzing liver biopsies from patients diagnosed with acute hepatic failure. Conclusion We suggest that under pathological conditions, ATP may act not only an immune system activator, but also as a paracrine direct cytotoxic DAMP through the dysregulation of Ca2+ homeostasis.

Details

Language :
English
Database :
OpenAIRE
Journal :
Cell Communication and Signaling : CCS
Accession number :
edsair.doi.dedup.....654d18ebfd8b69a22bc6c8d49171673c