13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Hypoxia truncates and constitutively activates the key cholesterol synthesis enzyme squalene monooxygenase

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Cholesterol synthesis is both energy- and oxygen-intensive, yet relatively little is known of the regulatory effects of hypoxia on pathway enzymes. We previously showed that the rate-limiting and first oxygen-dependent enzyme of the committed cholesterol synthesis pathway, squalene monooxygenase (SM), can undergo partial proteasomal degradation that renders it constitutively active. Here, we show hypoxia is a physiological trigger for this truncation, which occurs through a two-part mechanism: (1) increased targeting of SM to the proteasome via stabilization of the E3 ubiquitin ligase MARCHF6 and (2) accumulation of the SM substrate, squalene, which impedes the complete degradation of SM and liberates its truncated form. This preserves SM activity and downstream pathway flux during hypoxia. These results uncover a feedforward mechanism that allows SM to accommodate fluctuating substrate levels and may contribute to its widely reported oncogenic properties.

          eLife digest

          Cells need cholesterol to work properly but too much cholesterol is harmful and can contribute to atherosclerosis (narrowing of blood vessels), cancer and other diseases. Cells therefore carefully control the activity of the enzymes that are involved in making cholesterol, including an enzyme known as squalene monooxygenase.

          When the level of cholesterol in a cell rises, a protein called MARCHF6 adds molecules of ubiquitin to squalene monooxygenase. These molecules act as tags that direct the enzyme to be destroyed by a machine inside cells, known as the proteasome, thereby preventing further (unnecessary) production of cholesterol.

          Previous studies found that squalene monooxygenase is sometimes only partially broken down to make a shorter (truncated) form of the enzyme that is permanently active, even when the level of cholesterol in the cell is high. However, it was unclear what triggers this partial breakdown.

          The process of making cholesterol uses a lot of oxygen, yet many cancer cells thrive in tumours with low levels of oxygen. Here, Coates et al. used biochemical and cell biology approaches to study the effect of low oxygen levels on the activity of squalene monooxygenase in human cells. The experiments revealed that low oxygen levels trigger squalene monooxygenase to be partially degraded to make the truncated form of the enzyme.

          Firstly, MARCHF6 accumulates and adds ubiquitin to the enzyme to accelerate its delivery to the proteasome. Secondly, as the proteasome starts to degrade the enzyme, a build-up of squalene molecules impedes further breakdown of the enzyme. This mechanism preserves squalene monooxygenase activity when oxygen levels drop in cells, which may compensate for temporary oxygen shortfalls and allow cells to continue to make cholesterol.

          Squalene monooxygenase is overactive in individuals with a wide variety of diseases including fatty liver and prostate cancer. Drugs that block squalene monooxygenase activity have been shown to stop cancer cells from growing, but unfortunately these drugs are also toxic to mammals. These findings suggest that reducing the activity of squalene monooxygenase in more subtle ways, such as stopping it from being partially degraded, may be a more viable treatment strategy for cancer and other diseases associated with high levels of cholesterol.

          Related collections

          Most cited references69

          • Record: found
          • Abstract: not found
          • Article: not found

          Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genome engineering using the CRISPR-Cas9 system.

            Targeted nucleases are powerful tools for mediating genome alteration with high precision. The RNA-guided Cas9 nuclease from the microbial clustered regularly interspaced short palindromic repeats (CRISPR) adaptive immune system can be used to facilitate efficient genome engineering in eukaryotic cells by simply specifying a 20-nt targeting sequence within its guide RNA. Here we describe a set of tools for Cas9-mediated genome editing via nonhomologous end joining (NHEJ) or homology-directed repair (HDR) in mammalian cells, as well as generation of modified cell lines for downstream functional studies. To minimize off-target cleavage, we further describe a double-nicking strategy using the Cas9 nickase mutant with paired guide RNAs. This protocol provides experimentally derived guidelines for the selection of target sites, evaluation of cleavage efficiency and analysis of off-target activity. Beginning with target design, gene modifications can be achieved within as little as 1-2 weeks, and modified clonal cell lines can be derived within 2-3 weeks.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Analyzing real-time PCR data by the comparative C(T) method.

              Two different methods of presenting quantitative gene expression exist: absolute and relative quantification. Absolute quantification calculates the copy number of the gene usually by relating the PCR signal to a standard curve. Relative gene expression presents the data of the gene of interest relative to some calibrator or internal control gene. A widely used method to present relative gene expression is the comparative C(T) method also referred to as the 2 (-DeltaDeltaC(T)) method. This protocol provides an overview of the comparative C(T) method for quantitative gene expression studies. Also presented here are various examples to present quantitative gene expression data using this method.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing Editor
                Role: Senior Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                19 January 2023
                2023
                : 12
                : e82843
                Affiliations
                [1 ] School of Biotechnology and Biomolecular Sciences, UNSW Sydney ( https://ror.org/03r8z3t63) Sydney Australia
                [2 ] Prince of Wales Private Hospital ( https://ror.org/022arq532) Randwick Australia
                [3 ] Chris O’Brien Lifehouse ( https://ror.org/00qeks103) Camperdown Australia
                University of Texas Southwestern Medical Center ( https://ror.org/05byvp690) United States
                University of Cambridge ( https://ror.org/013meh722) United Kingdom
                University of Texas Southwestern Medical Center ( https://ror.org/05byvp690) United States
                University of Texas Southwestern Medical Center ( https://ror.org/05byvp690) United States
                Author information
                https://orcid.org/0000-0002-6506-5249
                https://orcid.org/0000-0002-6648-017X
                https://orcid.org/0000-0002-8482-6031
                https://orcid.org/0000-0002-4475-0116
                Article
                82843
                10.7554/eLife.82843
                9851614
                36655986
                05b72e0e-24b5-4887-bb66-3f19b16ad3ee
                © 2023, Coates et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 19 August 2022
                : 13 December 2022
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100015539, Australian Government;
                Award ID: Research Training Program Scholarship
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001773, University of New South Wales;
                Award ID: Scientia PhD Scholarship
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001104, RANZCOG Research Foundation;
                Award ID: Mary Elizabeth Courier Research Scholarship
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001171, Cancer Institute NSW;
                Award ID: Career Development Fellowship
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000923, Australian Research Council;
                Award ID: Grant DP170101178
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100009287, NSW Health;
                Award ID: Investigator Development Grant
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001171, Cancer Institute NSW;
                Award ID: 2021/CDF1120
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Biochemistry and Chemical Biology
                Custom metadata
                Hypoxic accumulation of squalene, the substrate of squalene monooxygenase, triggers its proteasomal truncation to a constitutively active variant that preserves downstream sterol synthesis.

                Life sciences
                squalene monooxygenase,cholesterol,hypoxia,proteasome,protein degradation,human
                Life sciences
                squalene monooxygenase, cholesterol, hypoxia, proteasome, protein degradation, human

                Comments

                Comment on this article