Elsevier

Gene

Volume 716, 20 October 2019, 144032
Gene

Research paper
Mitochondrial MTHFD isozymes display distinct expression, regulation, and association with cancer

https://doi.org/10.1016/j.gene.2019.144032Get rights and content

Highlights

  • Mammalian cells possess two mitochondrial MTHFD isozymes, MTHFD2 and MTHFD2L.

  • Both isozymes are present in normal and cancer cells, but MTHFD2 has in general higher expression.

  • MTHFD2 displays a more prominent response to growth factor stimulation.

  • There is no compensatory increase of MTHFD2L following inhibition of MTHFD2.

  • The therapeutic targeting of mitochondrial folate pathway should focus on the MTHFD2 isozyme.

Abstract

Mitochondrial folate metabolism is central to the generation of nucleotides, fuelling methylation reactions, and redox homeostasis. Uniquely among the reactions of the mitochondrial folate pathway, the key step of the oxidation of 5,10-methylene-tetrahydrofolate (CH2-THF) can be catalysed by two isozymes, MTHFD2 and MTHFD2L. The MTHFD2 enzyme has recently received considerable attention as an oncogenic enzyme upregulated in several tumour types, which is additionally required by cancer cells in vitro and in vivo. However, much less is currently known about MTHFD2L and its expression in cancer. In this study, we examine and compare the expression and regulation of the two mitochondrial MTHFD isozymes in normal human and cancer cells. We found that normal and cancer cells express both enzymes, although MTHFD2 has a much higher baseline expression. Unlike MTHFD2, the MTHFD2L isozyme does not show an association with proliferation and growth factor stimulation. In addition, we did not find evidence of a compensatory increase of MTHFD2L following suppression of its isozyme. This study supports that MTHFD2L is unlikely to have an important function in increased proliferation or cancer. Furthermore, therapeutic strategies aiming to block the mitochondrial folate pathway in cancer should focus on MTHFD2, with MTHFD2L being unlikely to be involved in the development of chemoresistance to targeting of its mitochondrial isozyme.

Introduction

Folate mediated one-carbon (1C) metabolism supports essential biological processes, which are often deregulated in cancer cells, such as biosynthesis of purines and thymidylate, homeostasis of amino acids (serine, glycine, methionine), methylation reactions, and regeneration of anti-oxidants. In eukaryotic cells, the folate 1C metabolism is compartmentalised into cytoplasmic, nuclear, and mitochondrial pathways (Tibbetts and Appling, 2010; Ducker and Rabinowitz, 2017). Experimental evidence supports that the mitochondrial compartment of the folate 1C pathway is the source for the vast majority of 1C units. These 1C units originate from molecules such as serine and glycine, and are processed through the folate pathway to generate formate units, which are eventually released into the cytosol (MacFarlane et al., 2008; Pike et al., 2010; Ducker et al., 2016).

Within the mitochondrial 1C pathway, each step is catalysed by a single enzyme, with the exception of the presence of two distinct bifunctional CH2-THF dehydrogenase/CH+-THF cyclohydrolase enzymes, MTHFD2 and MTHFD2L (Fig. 1). These two isozymes catalyse the conversion of CH2-THF into CHO-THF, which is the substrate for the production of formate through the action of MTHFD1L. Of these two isozymes, the most studied by far is MTHFD2. This enzyme was initially identified as a NAD+-dependent 5,10-CH2-THF dehydrogenase in 1985 (Mejia and MacKenzie, 1985). In recent years, a number of studies have implicated this enzyme in cancer. In a comparison of 14 different tumour/normal tissues, it was found that MTHFD2 is the most upregulated out of >1000 metabolic enzymes examined (Nilsson et al., 2014). Inhibition of MTHFD2 has been reported to affect proliferation and survival of various cancer cells, including lung (Nishimura et al., 2019), breast (Lehtinen et al., 2013; Koufaris et al., 2016a), and acute myeloid leukemia (Pikman et al., 2016). Consequently, MTHFD2 is considered a novel and promising therapeutic target, with a number of efforts in developing MTHFD2 inhibitors already reported (Fu et al., 2017; Gustafsson et al., 2017; Asai et al., 2018).

In contrast, the second mitochondrial MTHFD isozyme, MTHFD2L, has, so far, received much less attention. The Appling group has been the driver behind the identification and characterisation of this second mitochondrial MTHFD isozyme. By searching for novel vertebrate genes with sequence similarity to MTHFD1, they were able to identify and clone the MTHFD2L enzyme. It was also demonstrated that this enzyme possesses CH2-THF activity and that it is expressed in various adult tissues (Bolusani et al., 2011). In a subsequent study, the same group tested the isolated protein to verify that it had the same bifunctional catalytic capabilities as MTHFD2. Gene expression profiling in adult mice tissues found that the mRNA levels of MTHFD2L were much lower than MTHFD2, with the exception of the testes and spleen (Shin et al., 2014). Finally, in a more recent paper, this group showed that, besides possessing similar bifunctional enzymatic capabilities, both enzymes can use NAD+ or NADP+ as cofactors (Shin et al., 2017).

Despite the increasing attention given to the mitochondrial folate pathway in recent years, the interaction and roles of the MTHFD2/MTHFD2L dyad are still not understood. For instance, it has not yet been clarified whether cancer cells inverse the expression of the two isozymes, from low MTHFD2/high MTHFD2L to the reverse, and whether only expression of MTHFD2 is increased or whether both display increased levels. Another open issue is whether, upon suppression of MTHFD2, cells respond by increasing the levels of the MTHFD2L isozyme. Beyond biological interest, these questions are also of significant importance for optimising drug design strategies, e.g. whether chemicals targeting just MTHFD2 or both isozymes would be more efficient for targeting cancer cells.

In this study, we used a combination of experimental methods and the examination of publicly available data to compare, contrast, and define the regulation and expression of the two mitochondrial MTHFD isozymes. First, using this combinatorial approach, we show that the MTHFD2L isozyme is less dynamically regulated compared to MTHFD2, supporting a more baseline or housekeeping function. Second, cancer cells retain expression of both isozymes, but only MTHFD2 displays prominent upregulation in cancer. Third, MTHFD2L is not involved in the adaptive response of cells to suppression of MTHFD2. The novel insights generated by this project are important for understanding the regulation and function of the mammalian folate pathway.

Section snippets

Cell culture

Cell lines were grown and maintained in a 37 °C and 5% CO2 incubator in RPMI media with 5% FBS and penicillin/streptomycin, until the required experimental treatment. The MTHFD2 CRISPR−/− knockout cell lines were described previously (Ducker et al., 2016). For serum starvation experiments, 200,000 MCF7 cells were plated in six-well plates. The next day, the medium was removed, the cells were washed with PBS, and then serum-free RPMI-1640 medium was added for 48 h.

For transfections of siRNA

Expression of the two MTHFD isoforms in normal and cancer human cells

As a first step, we examined the mRNA levels of the MTHFD2 and MTHFD2L isozymes in normal human tissues by examining data from the GTex database (Suppl. Fig. 1&2). In the GTex dataset, the mRNA levels of MTHFD2L ranges from 6.8 transcripts per million (TPM) to 0.3 TPM in whole blood. For MTHFD2, the range is from 158 in EBV-transformed lymphocytes, to 1.2 in the liver. MTHFD2L mRNA was at higher levels in adult liver (4.6 vs 1.2) and comparable levels in some tissues (e.g. Brain cortex MTHFD2L

Discussion

In this study, we have, for the first time, performed an in-depth comparison of the expression profiles and response to growth factors, of the MTHFD2 and MTHFD2L isozymes. These analyses allowed us to draw the following conclusions: (1) Human cells maintain expression of both enzymes, with MTHFD2 generally having a higher expression in human tissues and cell lines; (2) The expression of MTHFD2L appears to be more stable, being less affected by carcinogenic transformation and growth factors; (3)

Author contribution

R.N. Conceptualization; Funding acquisition; Investigation; Methodology; Project administration; Resources; Writing - review & editing.

V.N. Data curation; Formal analysis; Investigation; Methodology; Resources; Writing - original draft; Writing - review & editing.

C.K. Conceptualization; Data curation; Formal analysis; Funding acquisition; Investigation; Methodology; Project administration; Resources; Supervision; Validation; Visualization; Writing - original draft; Writing - review & editing.

Declaration of competing interest

Authors declare no conflict of interest.

Acknowledgements

This work was supported by an Advanced Post-doctoral Fellowship from the University of Cyprus; Cancerfonden [CAN 2016/632]; and the Swedish Foundation for Strategic Research [FFL12-0220].

We would like to thank the Karaiskakio Foundation, Nicosia, Cyprus for the provision of materials and equipment. We also thank Dr. Christos Papaneophytou for his critical reading of the manuscript.

References (31)

  • X. Chen et al.

    Gene expression patterns in human liver cancers

    Mol. Biol. Cell

    (2002)
  • J. Cui et al.

    An integrated transcriptomic and computational analysis for biomarker identification in gastric cancer

    Nucleic Acids Res.

    (2011)
  • C. Fu et al.

    The natural product carolacton inhibits folate-dependent C1 metabolism by targeting FolD/MTHFD

    Nat. Commun.

    (2017)
  • N. Gustafsson Sheppard et al.

    The folate-coupled enzyme MTHFD2 is a nuclear protein and promotes cell proliferation

    Sci. Rep.

    (2015)
  • R. Gustafsson et al.

    Crystal structure of the emerging cancer target MTHFD2 in complex with a substrate-based inhibitor

    Cancer Res.

    (2017)
  • Cited by (17)

    View all citing articles on Scopus
    View full text