7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The TMEM189 gene encodes plasmanylethanolamine desaturase which introduces the characteristic vinyl ether double bond into plasmalogens

      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.

          Significance

          Although sequencing of the human genome was completed years ago, we still do not know about the physiological significance of thousands of predicted proteins, particularly of predicted membrane proteins. On the other hand, for approximately 100 human enzymes, no coding gene is known even though their enzymatic reaction has been well characterized. In this work, we assign one of those predicted membrane proteins (transmembrane protein 189; TMEM189) to one of the enzymatic reactions with an uncharacterized gene (plasmanylethanolamine desaturase). This enzyme catalyzes the final step in the biosynthesis of plasmalogens, an abundant class of glycerophospholipids that is depleted in such diseases as Alzheimer’s. Our findings enable interpretation of the previously characterized impaired growth phenotype of Tmem189-deficient mice.

          Abstract

          A significant fraction of the glycerophospholipids in the human body is composed of plasmalogens, particularly in the brain, cardiac, and immune cell membranes. A decline in these lipids has been observed in such diseases as Alzheimer’s and chronic obstructive pulmonary disease. Plasmalogens contain a characteristic 1- O-alk-1′-enyl ether (vinyl ether) double bond that confers special biophysical, biochemical, and chemical properties to these lipids. However, the genetics of their biosynthesis is not fully understood, since no gene has been identified that encodes plasmanylethanolamine desaturase (E.C. 1.14.99.19), the enzyme introducing the crucial alk-1′-enyl ether double bond. The present work identifies this gene as transmembrane protein 189 ( TMEM189). Inactivation of the TMEM189 gene in human HAP1 cells led to a total loss of plasmanylethanolamine desaturase activity, strongly decreased plasmalogen levels, and accumulation of plasmanylethanolamine substrates and resulted in an inability of these cells to form labeled plasmalogens from labeled alkylglycerols. Transient expression of TMEM189 protein, but not of other selected desaturases, recovered this deficit. TMEM189 proteins contain a conserved protein motif (pfam10520) with eight conserved histidines that is shared by an alternative type of plant desaturase but not by other mammalian proteins. Each of these histidines is essential for plasmanylethanolamine desaturase activity. Mice homozygous for an inactivated Tmem189 gene lacked plasmanylethanolamine desaturase activity and had dramatically lowered plasmalogen levels in their tissues. These results assign the TMEM189 gene to plasmanylethanolamine desaturase and suggest that the previously characterized phenotype of Tmem189-deficient mice may be caused by a lack of plasmalogens.

          Related collections

          Most cited references31

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

          Plasmalogens: biosynthesis and functions.

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

            X-ray Structure of a Mammalian Stearoyl-CoA Desaturase

            Stearoyl-CoA desaturase (SCD) is conserved in all eukaryotes and introduces the first double bond into saturated fatty acyl-CoAs 1–4 . Since the monounsaturated products of SCD are key precursors of membrane phospholipids, cholesterol esters, and triglycerides, SCD is pivotal in fatty acid metabolism. Humans have two SCD homologs (SCD1 and SCD5), while mice have four (SCD1–SCD4). SCD1-deficient mice do not become obese or diabetic when fed a high-fat diet because of improved lipid metabolic profiles and insulin sensitivity 5,6 . Thus, SCD1 is a pharmacological target in the treatment of obesity, diabetes, and other metabolic diseases 7 . SCD1 is an integral membrane protein located in the endoplasmic reticulum, and catalyzes the formation of a cis-double bond between the 9th and 10th carbons of stearoyl- or palmitoyl-CoA 8,9 . The reaction requires molecular oxygen, which is activated by a diiron center, and cytochrome b5, which regenerates the diiron center 10 . To better understand the structural basis of these characteristics of SCD function, we crystallized and solved the structure of mouse SCD1 bound to stearoyl-CoA at 2.6 Å resolution. The structure shows a novel fold comprising four transmembrane helices capped by a cytosolic domain, and a plausible pathway for lateral substrate access and product egress. The acyl chain of the bound stearoyl-CoA is enclosed in a tunnel buried in the cytosolic domain, and the geometry of the tunnel and configuration of the bound acyl chain provide a structural basis for the regioselectivity and stereospecificity of the desaturation reaction. The dimetal center is coordinated by a unique configuration of nine conserved histidine residues that implies a potentially novel metal center and mechanism for oxygen activation. The structure also illustrates a possible route for electron transfer from cytochrome b5 to the diiron center.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Eight histidine residues are catalytically essential in a membrane-associated iron enzyme, stearoyl-CoA desaturase, and are conserved in alkane hydroxylase and xylene monooxygenase.

              The eukaryotic fatty acid desaturases are iron-containing enzymes that catalyze the NAD-(P)H- and O2-dependent introduction of double bonds into methylene-interrupted fatty acyl chains. Examination of deduced amino acid sequences for the membrane desaturases from mammals, fungi, insects, higher plants, and cyanobacteria has revealed three regions of conserved primary sequence containing HX(3 or 4)H,HX(2 or 3)HH, and HX(2 or 3)HH. This motif is also present in the bacterial membrane enzymes alkane hydroxylase (omega-hydroxylase) and xylene monooxygenase. Hydropathy analyses indicate that these enzymes contain up to three long hydrophobic domains which would be long enough to span the membrane bilayer twice. The conserved His-containing regions have a consistent positioning with respect to these potential membrane spanning domains. Taken together, these observations suggest that the membrane fatty acid desaturases and hydrocarbon hydroxylases have a related protein fold, possibly arising from a common ancestral origin. In order to examine the functional role of these conserved His residues, we have made use of the ability of the rat delta 9 desaturase gene to complement a yeast strain deficient in the delta 9 desaturase gene function (ole1). By site-directed mutagenesis, eight conserved His residues in the rat delta 9 desaturase were individually converted to Ala. Each His-->Ala mutation failed to complement the yeast ole1 mutant. In contrast, mutation of three nonconserved flanking His residues or a partially conserved Arg residue within the conserved motif to Ala allowed for complementation of the ole1 phenotype.(ABSTRACT TRUNCATED AT 250 WORDS)
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                7 April 2020
                24 March 2020
                24 March 2020
                : 117
                : 14
                : 7792-7798
                Affiliations
                [1] aInstitute of Biological Chemistry, Biocenter, Medical University of Innsbruck , A-6020 Innsbruck, Austria;
                [2] bInstitute of Human Genetics, Medical University of Innsbruck , A-6020 Innsbruck, Austria;
                [3] cUniversity Clinic for Anesthesiology and General Intensive Care Medicine, Medical University of Innsbruck , A-6020 Innsbruck, Austria;
                [4] dInstitute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck , A-6020 Innsbruck, Austria;
                [5] eDepartment of Physiology and Biophysics, Boston University School of Medicine , Boston, MA 02118;
                [6] fInstitute of Genetics and Genomics, University of Geneva , 1211 Geneva 4, Switzerland;
                [7] gDepartment of Pathobiology of the Nervous System, Medical University of Vienna , 1090 Vienna, Austria
                Author notes
                1To whom correspondence may be addressed. Email: ernst.r.werner@ 123456i-med.ac.at or katrin.watschinger@ 123456i-med.ac.at .

                Edited by Benjamin F. Cravatt, Scripps Research Institute, La Jolla, CA, and approved February 26, 2020 (received for review October 7, 2019)

                Author contributions: E.R.W., M.A.K., G.G., G.W.-F., N.H., J.B., and K.W. designed research; E.R.W., M.A.K., S.S., K.L., J.K., M.H., S.C., N.H., and K.W. performed research; R.A.Z. contributed new reagents/analytic tools; and E.R.W. wrote the paper.

                Author information
                http://orcid.org/0000-0003-1948-3391
                http://orcid.org/0000-0002-8654-9920
                http://orcid.org/0000-0002-6375-5393
                http://orcid.org/0000-0003-3260-9139
                http://orcid.org/0000-0003-2962-125X
                http://orcid.org/0000-0002-6560-0993
                http://orcid.org/0000-0003-0182-2658
                http://orcid.org/0000-0002-1122-8444
                Article
                201917461
                10.1073/pnas.1917461117
                7149458
                32209662
                0369e05f-9930-4f81-a717-1c8b2eaaa05d
                Copyright © 2020 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution License 4.0 (CC BY).

                History
                Page count
                Pages: 7
                Funding
                Funded by: Austrian Science Fund (FWF) 501100002428
                Award ID: P29551
                Award Recipient : Ernst R Werner Award Recipient : Johannes Berger Award Recipient : Katrin Watschinger
                Funded by: Austrian Science Fund (FWF) 501100002428
                Award ID: P30800
                Award Recipient : Ernst R Werner Award Recipient : Johannes Berger Award Recipient : Katrin Watschinger
                Funded by: Austrian Science Fund (FWF) 501100002428
                Award ID: I2738
                Award Recipient : Ernst R Werner Award Recipient : Johannes Berger Award Recipient : Katrin Watschinger
                Categories
                Biological Sciences
                Biochemistry

                plasmalogen,transmembrane protein 189,plasmanylethanolamine desaturase,ether lipid

                Comments

                Comment on this article