A critical role for heme synthesis and succinate in the regulation of pluripotent states transitions

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    In their current study, Detraux D and colleagues provide some evidence suggesting a role for heme biosynthesis on FGF-ERK and TGF beta signalling and exit from naïve pluripotency, and in controlling the 2-cell-like cell state. The observations provided by the authors are interesting and potentially relevant in the field of pluripotent cell state transitions.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Using embryonic stem cells (ESCs) in regenerative medicine or in disease modeling requires a complete understanding of these cells. Two main distinct developmental states of ESCs have been stabilized in vitro, a naïve pre-implantation stage and a primed post-implantation stage. Based on two recently published CRISPR-Cas9 knockout functional screens, we show here that the exit of the naïve state is impaired upon heme biosynthesis pathway blockade, linked in mESCs to the incapacity to activate MAPK- and TGFβ-dependent signaling pathways after succinate accumulation. In addition, heme synthesis inhibition promotes the acquisition of 2 cell-like cells in a heme-independent manner caused by a mitochondrial succinate accumulation and leakage out of the cell. We further demonstrate that extracellular succinate acts as a paracrine/autocrine signal, able to trigger the 2C-like reprogramming through the activation of its plasma membrane receptor, SUCNR1. Overall, this study unveils a new mechanism underlying the maintenance of pluripotency under the control of heme synthesis.

Article activity feed

  1. eLife assessment

    In their current study, Detraux D and colleagues provide some evidence suggesting a role for heme biosynthesis on FGF-ERK and TGF beta signalling and exit from naïve pluripotency, and in controlling the 2-cell-like cell state. The observations provided by the authors are interesting and potentially relevant in the field of pluripotent cell state transitions.

  2. Reviewer #1 (Public Review):

    In the manuscript the authors identify new players regulating cell state transitions. They show that heme biosynthesis is required for the naïve-to-primed pluripotency transition. In particular, they provide a link between heme biosynthesis inhibition and failure to activate TGFβ and MAPK pathways, two crucial regulators of the exit from the naïve pluripotent state. Heme biosynthesis inhibition increases the percentage of 2CLCs within the mESC population. The authors further show that this increased level of 2CLCs depends on the accumulation of succinate in non-mitochondrial cell compartments as a consequence of heme biosynthesis inhibition. Based on experiments using chemical inhibitors, the authors conclude that succinate acts in a paracrine and autocrine manner to enhance reprogramming of mESC into 2CLCs.

    The observations provided by the authors are interesting and potentially relevant in the field of pluripotent cell state transitions. However, in the present state, neither the role of heme biosynthesis on FGF-ERK and TGF beta signalling and exit from naïve pluripotency nor the role of heme biosynthesis in controlling the 2CLC state are clear.

    Below I list my main concerns:

    1. The authors claim that the heme metabolic pathway is important for the naïve-to-primed transition. Interfering with its proper function appears to have developmental consequences. However, it is unclear how exactly this pathway is regulated during the exit from naïve pluripotency in WT cells. Hence the physiological relevance remains unclear. Are the levels of heme itself, or the mentioned 7 enzymes of the heme pathway regulated during differentiation?

    2. The claim that "the roles of this [heme] biosynthetic pathway and this metabolite have never been studied in the context of pluripotency" is a bit misleading. It has been reported that HO1 is regulated by Oct4 (https://doi.org/10.1002/1873-3468.14138).

    3. The link between heme biosynthesis and the TGFβ and MAPK pathways remains unclear. Is there any evidence for a direct link, or are these two observations simply linked through an altered cell state? Without further experiments it remains unclear whether the lack of proper Tgf beta and Fgf-ERK signalling activation are cause or consequence of the observed differentiation defects. Results must be discussed with this limitation in mind.

    4. The fact that MEKi did not recapitulate the phenotype of SA treatment to prevent EpiLC differentiation should already be clarified in the results section. Moreover, the fact that SMAD inhibition seems to delay downregulation of naïve markers more than SA treatment, and the fact that SMAD inhibition combined with MEK inhibition seems weaker than SMAD inhibition alone seem counterintuitive and needs explanation. Can the authors attempt to titrate pathway inhibition to a similar level as observed in heme pathway deficient ESCs? Furthermore, can the differentiation defect be rescued upon overstimulation of Fgf-ERK and TGF beta to reach WT levels?

    5. To better characterize the direct effect of heme biosynthesis inhibition it is necessary to in depth analyse any possible cell proliferation, viability of cell cycle defects after SA (or AA5) treatment. If there is an impact on cellular health, this needs to be reported and taken into careful consideration when interpreting results.

    6. The authors claim that SA pre-treatment of mESC is able to enhance their differentiation ability into throphoblast like cells; however, they do not show statistically significant differences in terms of throphoblast expression markers between SA-treated and control cells (Supplemental Fig.2d). Furthermore, the variance of measurements in 2iL are not shown. Expression levels in TS cells or in trophoblast tissue must be used as control to judge the effect size. 2iL cells do also generate cells similar in morphology to 2iL+SA cells (Suppl Fig. 2b). Should these also be giant cells; this would be very surprising. Together, this makes drawing conclusions from these experiments impossible. If the statement that SA treatment expands the lineage potential of ESCs is made, it will need to be supported by appropriate and statistically strong data. A mere increase in some marker genes (which are not really specific for the TE lineage but also expressed in embryonic tissues) and statements based on morphology are no good support for this hypothesis.

    7. What is the reason for calling the increase in 2C like cells 2C-like reprogramming? Is there any evidence that this is indeed a reprogramming event? There is no evidence for disruption of heme biosynthesis directly instructing cells to take on a 2CLC state, there is simply an increase in the Zscan4 expressing population, for a reason that remains unclear.

    8. Addition of AA5 or SA results in absolutely striking changes in the epigenetic state of ESCs. H3K4me3, H3K27me3 and H3K9me3 together with 5mC levels appear drastically increased based on IF images in Supp. Fig. 5a. I wonder how physiological these levels are. How much data directly meaningful for 'normal' differentiation can be obtained from such a massively perturbed system? This needs to be appropriately acknowledged and discussed.