Abstract

Steroidogenic acute regulatory (StAR) proteins in steroidogenic cells are implicated in the delivery of cholesterol (Ch) from internal or external sources to mitochondria (Mito) for initiation of steroid hormone synthesis. In this study, we tested the hypothesis that under oxidative stress, StAR-mediated trafficking of redox-active cholesterol hydroperoxides (ChOOHs) can result in site-specific Mito damage and dysfunction. Steroidogenic stimulation of mouse MA-10 Leydig cells with dibutyryl-cAMP (Bt2cAMP) resulted in strong expression of StarD1 and StarD4 proteins over insignificant levels in nonstimulated controls. During incubation with the ChOOH 3β-hydroxycholest-5-ene-7α-hydroperoxide (7α-OOH) in liposomes, stimulated cells took up substantially more hydroperoxide in Mito than controls, with a resulting loss of membrane potential (ΔΨm) and ability to drive progesterone synthesis. 7α-OOH uptake and ΔΨm loss were greatly reduced by StarD1 knockdown, thus establishing the role of this protein in 7α-OOH delivery. Moreover, 7α-OOH was substantially more toxic to stimulated than nonstimulated cells, the former dying mainly by apoptosis and the latter dying by necrosis. Importantly, tert-butyl hydroperoxide, which is not a StAR protein ligand, was equally toxic to stimulated and nonstimulated cells. These findings support the notion that like Ch itself, 7α-OOH can be transported to/into Mito of steroidogenic cells by StAR proteins and therein induce free radical damage, which compromises steroid hormone synthesis.

Highlights

  • Steroidogenic acute regulatory (StAR) proteins in steroidogenic cells transport cholesterol to/into mitochondria

  • During incubation with the cholesterol hydroperoxides (ChOOHs) 3␤-hydroxycholest5-ene-7␣-hydroperoxide (7␣-OOH) in liposomes, stimulated cells took up substantially more hydroperoxide in Mito than controls, with a resulting loss of membrane potential (⌬⌿m) and ability to drive progesterone synthesis. 7␣-OOH uptake and ⌬⌿m loss were greatly reduced by StarD1 knockdown, establishing the role of this protein in 7␣-OOH delivery

  • Consistent with this, we observed a strong induction of immunodetectable StarD1 (ϳ27 kDa) in MA-10 cells after treating with 1 mM Bt2cAMP in serum-free Dulbecco’s modified Eagle’s/Ham’s nutrient F12 (DME/F12) medium (Fig. 1A)

Read more

Summary

Background

StAR proteins in steroidogenic cells transport cholesterol to/into mitochondria. Under oxidative stress, StARs might deliver damaging cholesterol hydroperoxides (ChOOHs). Results: Steroidogenic activation of MA-10 Leydig cells results in StarD1/D4 expression, ChOOH delivery to mitochondria, membrane potential loss, and reduced progesterone output. Tert-butyl hydroperoxide, which is not a StAR protein ligand, was toxic to stimulated and nonstimulated cells These findings support the notion that like Ch itself, 7␣-OOH can be transported to/into Mito of steroidogenic cells by StAR proteins and therein induce free radical damage, which compromises steroid hormone synthesis. We showed that transfer of 7␣-hydroperoxycholesterol (7␣-OOH) from liposomes to isolated Mito was strongly enhanced by recombinant StarD4 and that this induced Mito peroxidative damage and loss of membrane potential [24] This was the first reported evidence for a StAR family protein acting in this manner. We report that steroidogenic activation of mouse MA-10 Leydig cells, as evidenced by StAR protein expression and progesterone synthesis, makes these cells remarkably more sensitive to redox damage and dysfunction by Mito-targeted 7␣-OOH

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call