NAMPT-derived NAD+ fuels PARP1 to promote skin inflammation through parthanatos cell death

PLoS Biol. 2021 Nov 8;19(11):e3001455. doi: 10.1371/journal.pbio.3001455. eCollection 2021 Nov.

Abstract

Several studies have revealed a correlation between chronic inflammation and nicotinamide adenine dinucleotide (NAD+) metabolism, but the precise mechanism involved is unknown. Here, we report that the genetic and pharmacological inhibition of nicotinamide phosphoribosyltransferase (Nampt), the rate-limiting enzyme in the salvage pathway of NAD+ biosynthesis, reduced oxidative stress, inflammation, and keratinocyte DNA damage, hyperproliferation, and cell death in zebrafish models of chronic skin inflammation, while all these effects were reversed by NAD+ supplementation. Similarly, genetic and pharmacological inhibition of poly(ADP-ribose) (PAR) polymerase 1 (Parp1), overexpression of PAR glycohydrolase, inhibition of apoptosis-inducing factor 1, inhibition of NADPH oxidases, and reactive oxygen species (ROS) scavenging all phenocopied the effects of Nampt inhibition. Pharmacological inhibition of NADPH oxidases/NAMPT/PARP/AIFM1 axis decreased the expression of pathology-associated genes in human organotypic 3D skin models of psoriasis. Consistently, an aberrant induction of NAMPT and PARP activity, together with AIFM1 nuclear translocation, was observed in lesional skin from psoriasis patients. In conclusion, hyperactivation of PARP1 in response to ROS-induced DNA damage, fueled by NAMPT-derived NAD+, mediates skin inflammation through parthanatos cell death.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis Inducing Factor / metabolism
  • Cell Nucleus / drug effects
  • Cell Nucleus / metabolism
  • Cell Proliferation / drug effects
  • DNA Damage
  • Disease Models, Animal
  • Gene Expression Regulation / drug effects
  • Inflammation / genetics
  • Inflammation / pathology*
  • Keratinocytes / drug effects
  • Keratinocytes / metabolism
  • Keratinocytes / pathology
  • Larva / metabolism
  • NAD / metabolism*
  • NADPH Oxidases / antagonists & inhibitors
  • NADPH Oxidases / metabolism
  • Nicotinamide Phosphoribosyltransferase / antagonists & inhibitors
  • Nicotinamide Phosphoribosyltransferase / metabolism*
  • Oxidative Stress / drug effects
  • Oxidative Stress / genetics
  • Parthanatos* / drug effects
  • Parthanatos* / genetics
  • Poly Adenosine Diphosphate Ribose / metabolism
  • Poly(ADP-ribose) Polymerase Inhibitors / pharmacology
  • Poly(ADP-ribose) Polymerases / metabolism*
  • Proteinase Inhibitory Proteins, Secretory / deficiency
  • Proteinase Inhibitory Proteins, Secretory / metabolism
  • Psoriasis / genetics
  • Psoriasis / pathology
  • Reactive Oxygen Species / metabolism
  • Skin / pathology*
  • Zebrafish
  • Zebrafish Proteins / deficiency
  • Zebrafish Proteins / metabolism

Substances

  • AIFM1 protein, human
  • Apoptosis Inducing Factor
  • Poly(ADP-ribose) Polymerase Inhibitors
  • Proteinase Inhibitory Proteins, Secretory
  • Reactive Oxygen Species
  • Zebrafish Proteins
  • spint1a protein, zebrafish
  • NAD
  • Poly Adenosine Diphosphate Ribose
  • NADPH Oxidases
  • Nicotinamide Phosphoribosyltransferase
  • Poly(ADP-ribose) Polymerases

Grants and funding

This work was supported by grants BIO2017-84702-R and PID2020-113660RB-I00 to VM and PhD fellowship to FJMN funded by MCIN/AEI/10.13039/501100011033 and European Regional Development Funds, grant 20793/PI/18 to VM funded by Fundación Séneca-Murcia, and contracts to ABPO, DGM and FJMM funded by Universidad de Murcia. The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.