Sphingosine-1-phosphate lyase mutations cause primary adrenal insufficiency and steroid-resistant nephrotic syndrome

Rathi Prasad, Irene Hadjidemetriou, Avinaash Maharaj, Eirini Meimaridou, Federica Buonocore, Moin Saleem, Jenny Hurcombe, Agnieszka Bierzynska, Eliana Barbagelata, Ignacio Bergadá, Hamilton Cassinelli, Urmi Das, Ruth Krone, Bulent Hacihamdioglu, Erkan Sari, Ediz Yesilkaya, Helen L Storr, Maria Clemente, Monica Fernandez-Cancio, Nuria Camats, Nanik Ram, John C Achermann, Paul P Van Veldhoven, Leonardo Guasti, Debora Braslavsky, Tulay Guran, Louise A Metherell, Rathi Prasad, Irene Hadjidemetriou, Avinaash Maharaj, Eirini Meimaridou, Federica Buonocore, Moin Saleem, Jenny Hurcombe, Agnieszka Bierzynska, Eliana Barbagelata, Ignacio Bergadá, Hamilton Cassinelli, Urmi Das, Ruth Krone, Bulent Hacihamdioglu, Erkan Sari, Ediz Yesilkaya, Helen L Storr, Maria Clemente, Monica Fernandez-Cancio, Nuria Camats, Nanik Ram, John C Achermann, Paul P Van Veldhoven, Leonardo Guasti, Debora Braslavsky, Tulay Guran, Louise A Metherell

Abstract

Primary adrenal insufficiency is life threatening and can present alone or in combination with other comorbidities. Here, we have described a primary adrenal insufficiency syndrome and steroid-resistant nephrotic syndrome caused by loss-of-function mutations in sphingosine-1-phosphate lyase (SGPL1). SGPL1 executes the final decisive step of the sphingolipid breakdown pathway, mediating the irreversible cleavage of the lipid-signaling molecule sphingosine-1-phosphate (S1P). Mutations in other upstream components of the pathway lead to harmful accumulation of lysosomal sphingolipid species, which are associated with a series of conditions known as the sphingolipidoses. In this work, we have identified 4 different homozygous mutations, c.665G>A (p.R222Q), c.1633_1635delTTC (p.F545del), c.261+1G>A (p.S65Rfs*6), and c.7dupA (p.S3Kfs*11), in 5 families with the condition. In total, 8 patients were investigated, some of whom also manifested other features, including ichthyosis, primary hypothyroidism, neurological symptoms, and cryptorchidism. Sgpl1-/- mice recapitulated the main characteristics of the human disease with abnormal adrenal and renal morphology. Sgpl1-/- mice displayed disrupted adrenocortical zonation and defective expression of steroidogenic enzymes as well as renal histology in keeping with a glomerular phenotype. In summary, we have identified SGPL1 mutations in humans that perhaps represent a distinct multisystemic disorder of sphingolipid metabolism.

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1. Pedigrees of kindreds 1 to…
Figure 1. Pedigrees of kindreds 1 to 5 where all affected individuals manifested PAI, with or without SRNS, and were positive for mutations in SGPL1.
Black symbols indicate individuals with PAI alone, half-filled in green indicate those who additionally had steroid-resistant nephrotic syndrome, and green indicate those with SRNS alone. All affected individuals were homozygous for the indicated mutations (patients sequenced have been numbered from 1 to 8), and parents were heterozygous (those sequenced denoted by gray symbols). Mutations for patients 2, 3, and 5 were identified by WES and the remainder by Sanger sequencing of SGPL1.
Figure 2. p.R222Q and p.F545del mutations affect…
Figure 2. p.R222Q and p.F545del mutations affect highly conserved areas in SGPL1 and are loss of function, resulting in proteins with reduced lyase activity.
(A) SGPL1 regulates flow of the sphingolipid biochemical intermediates (in green) and carries out the final degradation step in the pathway. (B) Partial alignment of SGPL1 protein sequences, generated by Clustal Omega (48), showing conservation of arginine (R) at position 222 and phenylalanine (F) at position 545, highlighted in yellow, with numbering relative to human sequence. For all but the most distant organisms, these amino acids are conserved. Alignment source accession numbers from ENSEMBL are as follows: Homo sapiens, human, ENSP00000362298; Mus musculus, mouse, ENSMUSP00000112975; Rattus norvegicus, rat, ENSRNOP00000070983; Tetraodon nigroviridis, pufferfish, ENSTNIP00000016065; Xenopus laevis, clawed frog, ENSXETP00000017960; Ciona intestinalis, sea squirt, ENSCINP00000002369; Drosophila melanogaster, fruit fly, FBpp0086158; Caenorhabditis elegans, nematode, B0222.4; and Saccharomyces cerevisiae, yeast, YDR294C. Sequence conservation is beneath the alignment. Asterisks indicate total conservation; colons indicate partial conservation. (C) SGPL1 activities were measured in lysates of Sgpl1–/– mouse fibroblasts. **P < 0.01, 2-tailed Student’s t test (n = 3). (D) Lysates of Sgpl1–/– mouse fibroblasts expressing WT or the mutant SGPL1 (25 μg protein/lane) were analyzed by immunoblotting for the presence of the FLAG-tagged protein (representative image, n = 3). (E) SGPL1 mRNA expression in a human tissue panel, analysis using the 2–ΔΔCT algorithm (n = 3). S intestine, small intestine.
Figure 3. Adrenals from Sgpl1 –/– mice…
Figure 3. Adrenals from Sgpl1–/– mice show histological abnormalities, and SGPL1 is expressed in human adrenals.
(A and B) Adrenals from Sgpl1–/– mice show histological abnormalities. (A) H&E staining of Sgpl1+/+ and Sgpl1–/– adrenals. Note the less defined morphological zonation in the Sgpl1–/– adrenals compared with that from Sgpl1+/+ mice. Moreover, the characteristic lipid droplets found in the ZF (arrowheads in top-right panel) and visible as large areas in the cytoplasm devoid of eosin staining (as lipids are extracted during the paraffin-embedding procedure) are strongly reduced in Sgpl1–/– adrenals (n = 3). Cap, capsule. Scale bars: 100 μm (left); 25 μm (middle); 5 μm (right). (B) CYP11A1 and CYP11B2 expression in Sgpl1+/+ and Sgpl1–/– adrenals. CYP11A1 staining in Sgpl1–/– adrenals is less prominent compared with Sgpl1+/+, while the characteristic patchy expression of aldosterone synthase (CYP11B2) is lost in Sgpl1–/– adrenals (n = 3). Scale bars: 100 μm (top); 25 μm (bottom). (CE) Expression of SGPL1 in human adrenals. (C) Western blotting of lysates from human adrenal, HEK293, cells and HEK293 cells overexpressing SGPL1 probed with anti-SGPL1 antibody (representative image of n = 3). (D) SGPL1 expression in the HFA at 19 and 22 Carnegie (Carn) stage as well as at 18 weeks showing widespread expression (n = 1 each). FZ, fetal zone; DZ, definitive zone. Scale bars: 100 μm. (E) SGPL1 expression in the human adult adrenal (n = 3). Note the stronger expression of SGPL1 in the ZR compared with ZG and ZF, while the capsule and medulla (M) are negative. Scale bars: 100 μm (left panel); 25 μm (right 3 panels).
Figure 4. Histological features of the glomeruli.
Figure 4. Histological features of the glomeruli.
(AC) H&E staining of Sgpl1+/+ kidney showing normal cortical histology (A) and glomeruli with open capillary loops and normal cellularity (B and C, yellow arrowhead). The kidneys from Sgpl1–/– mice (EG) have mild mesangial hypercellularity with glomerular hypertrophy (F and G, yellow arrowhead) and large protein casts in the tubules (white arrows). (D and H) Masson’s trichrome stain. Kidneys from Sgpl1–/– mice (H) show increased glomerular fibrosis (red stain) compared with Sgpl1+/+ (D). n = 3 in all cases. Scale bars: 100 μm (A and E); 25 μm (BD and FH).

References

    1. Charmandari E, Nicolaides NC, Chrousos GP. Adrenal insufficiency. Lancet. 2014;383(9935):2152–2167. doi: 10.1016/S0140-6736(13)61684-0.
    1. Malikova J, Flück CE. Novel insight into etiology, diagnosis and management of primary adrenal insufficiency. Horm Res Paediatr. 2014;82(3):145–157. doi: 10.1159/000363107.
    1. Chan LF, Campbell DC, Novoselova TV, Clark AJ, Metherell LA. Whole-exome sequencing in the differential diagnosis of primary adrenal insufficiency in children. Front Endocrinol (Lausanne) 2015;6:113.
    1. Clark AJ, McLoughlin L, Grossman A. Familial glucocorticoid deficiency associated with point mutation in the adrenocorticotropin receptor. Lancet. 1993;341(8843):461–462. doi: 10.1016/0140-6736(93)90208-X.
    1. Hughes CR, et al. MCM4 mutation causes adrenal failure, short stature, and natural killer cell deficiency in humans. J Clin Invest. 2012;122(3):814–820. doi: 10.1172/JCI60224.
    1. Meimaridou E, et al. Mutations in NNT encoding nicotinamide nucleotide transhydrogenase cause familial glucocorticoid deficiency. Nat Genet. 2012;44(7):740–742. doi: 10.1038/ng.2299.
    1. Metherell LA, et al. Mutations in MRAP, encoding a new interacting partner of the ACTH receptor, cause familial glucocorticoid deficiency type 2. Nat Genet. 2005;37(2):166–170. doi: 10.1038/ng1501.
    1. Prasad R, et al. Thioredoxin Reductase 2 (TXNRD2) mutation associated with familial glucocorticoid deficiency (FGD) J Clin Endocrinol Metab. 2014;99(8):E1556–E1563. doi: 10.1210/jc.2013-3844.
    1. Ram N, Asghar A, Islam N. A case report: Familial glucocorticoid deficiency associated with familial focal segmental glomerulosclerosis. BMC Endocr Disord. 2012;12:32.
    1. Aguilar A, Saba JD. Truth and consequences of sphingosine-1-phosphate lyase. Adv Biol Regul. 2012;52(1):17–30. doi: 10.1016/j.advenzreg.2011.09.015.
    1. Serra M, Saba JD. Sphingosine 1-phosphate lyase, a key regulator of sphingosine 1-phosphate signaling and function. Adv Enzyme Regul. 2010;50(1):349–362. doi: 10.1016/j.advenzreg.2009.10.024.
    1. Thomas AC, et al. Mutations in SNX14 cause a distinctive autosomal-recessive cerebellar ataxia and intellectual disability syndrome. Am J Hum Genet. 2014;95(5):611–621. doi: 10.1016/j.ajhg.2014.10.007.
    1. Borowsky AD, et al. Sphingosine-1-phosphate lyase expression in embryonic and adult murine tissues. J Lipid Res. 2012;53(9):1920–1931. doi: 10.1194/jlr.M028084.
    1. Van Veldhoven PP, Gijsbers S, Mannaerts GP, Vermeesch JR, Brys V. Human sphingosine-1-phosphate lyase: cDNA cloning, functional expression studies and mapping to chromosome 10q22(1) Biochim Biophys Acta. 2000;1487(2-3):128–134.
    1. Van Veldhoven PP, Mannaerts GP. Sphingosine-phosphate lyase. Adv Lipid Res. 1993;26:69–98.
    1. Platt FM. Sphingolipid lysosomal storage disorders. Nature. 2014;510(7503):68–75. doi: 10.1038/nature13476.
    1. Merscher S, Fornoni A. Podocyte pathology and nephropathy - sphingolipids in glomerular diseases. Front Endocrinol (Lausanne) 2014;5:127.
    1. Frankenburg WK, Dodds J, Archer P, Shapiro H, Bresnick B. The Denver II; a major revision and restandardization of the Denver Developmental Screening Test. Pediatrics. 1992;89(1):91–97.
    1. Zhou J, Saba JD. Identification of the first mammalian sphingosine phosphate lyase gene and its functional expression in yeast. Biochem Biophys Res Commun. 1998;242(3):502–507. doi: 10.1006/bbrc.1997.7993.
    1. Van Veldhoven PP. Sphingosine 1-phosphate lyase deficient mice [abstract] Chem Phys Lipids. 2005;136(2):164–165.
    1. Schmahl J, Raymond CS, Soriano P. PDGF signaling specificity is mediated through multiple immediate early genes. Nat Genet. 2007;39(1):52–60. doi: 10.1038/ng1922.
    1. Vogel P, et al. Incomplete inhibition of sphingosine 1-phosphate lyase modulates immune system function yet prevents early lethality and non-lymphoid lesions. PLoS ONE. 2009;4(1):e4112. doi: 10.1371/journal.pone.0004112.
    1. Schmahl J, Rizzolo K, Soriano P. The PDGF signaling pathway controls multiple steroid-producing lineages. Genes Dev. 2008;22(23):3255–3267. doi: 10.1101/gad.1723908.
    1. Beuschlein F, et al. SF-1, DAX-1, and acd: molecular determinants of adrenocortical growth and steroidogenesis. Endocr Res. 2002;28(4):597–607. doi: 10.1081/ERC-120016972.
    1. Schümann J, et al. Reduced activity of sphingosine-1-phosphate lyase induces podocyte-related glomerular proteinuria, skin irritation, and platelet activation. Toxicol Pathol. 2015;43(5):694–703. doi: 10.1177/0192623314565650.
    1. Li QL, Ni J, Bian SL, Yao LC, Zhu H, Zhang W. Inhibition of steroidogenesis and induction of apoptosis in rat luteal cells by cell-permeable ceramide in vitro. Sheng Li Xue Bao. 2001;53(2):142–146.
    1. Meroni SB, Pellizzari EH, Cánepa DF, Cigorraga SB. Possible involvement of ceramide in the regulation of rat Leydig cell function. J Steroid Biochem Mol Biol. 2000;75(4-5):307–313. doi: 10.1016/S0960-0760(00)00188-6.
    1. Urs AN, Dammer E, Kelly S, Wang E, Merrill AH, Sewer MB. Steroidogenic factor-1 is a sphingolipid binding protein. Mol Cell Endocrinol. 2007;265-266:174–178. doi: 10.1016/j.mce.2006.12.016.
    1. Lucki NC, Li D, Sewer MB. Sphingosine-1-phosphate rapidly increases cortisol biosynthesis and the expression of genes involved in cholesterol uptake and transport in H295R adrenocortical cells. Mol Cell Endocrinol. 2012;348(1):165–175. doi: 10.1016/j.mce.2011.08.003.
    1. Bektas M, et al. Sphingosine 1-phosphate lyase deficiency disrupts lipid homeostasis in liver. J Biol Chem. 2010;285(14):10880–10889. doi: 10.1074/jbc.M109.081489.
    1. Hagen N, Hans M, Hartmann D, Swandulla D, van Echten-Deckert G. Sphingosine-1-phosphate links glycosphingolipid metabolism to neurodegeneration via a calpain-mediated mechanism. Cell Death Differ. 2011;18(8):1356–1365. doi: 10.1038/cdd.2011.7.
    1. Uchida Y, et al. Epidermal sphingomyelins are precursors for selected stratum corneum ceramides. J Lipid Res. 2000;41(12):2071–2082.
    1. Holleran WM, et al. Consequences of beta-glucocerebrosidase deficiency in epidermis. Ultrastructure and permeability barrier alterations in Gaucher disease. J Clin Invest. 1994;93(4):1756–1764. doi: 10.1172/JCI117160.
    1. Gharib H, Hodgson SF, Gastineau CF, Scholz DA, Smith LA. Reversible hypothyroidism in Addison’s disease. Lancet. 1972;2(7780):734–736.
    1. Jeffcoate WJ, Davis JR. Reversible hypothyroidism in adrenal insufficiency. Br Med J (Clin Res Ed) 1982;285(6342):651–652.
    1. Topliss DJ, White EL, Stockigt JR. Significance of thyrotropin excess in untreated primary adrenal insufficiency. J Clin Endocrinol Metab. 1980;50(1):52–56.
    1. Feinstein EI, Kaptein EM, Nicoloff JT, Massry SG. Thyroid function in patients with nephrotic syndrome and normal renal function. Am J Nephrol. 1982;2(2):70–76. doi: 10.1159/000166587.
    1. Genter MB, et al. Microarray-based discovery of highly expressed olfactory mucosal genes: potential roles in the various functions of the olfactory system. Physiol Genomics. 2003;16(1):67–81. doi: 10.1152/physiolgenomics.00117.2003.
    1. Weber C, Krueger A, Münk A, Bode C, Van Veldhoven PP, Gräler MH. Discontinued postnatal thymocyte development in sphingosine 1-phosphate-lyase-deficient mice. J Immunol. 2009;183(7):4292–4301. doi: 10.4049/jimmunol.0901724.
    1. Bagdanoff JT, et al. Inhibition of sphingosine 1-phosphate lyase for the treatment of rheumatoid arthritis: discovery of (E)-1-(4-((1R,2S,3R)-1,2,3,4-tetrahydroxybutyl)-1H-imidazol-2-yl)ethanone oxime (LX2931) and (1R,2S,3R)-1-(2-(isoxazol-3-yl)-1H-imidazol-4-yl)butane-1,2,3,4-tetraol (LX2932) J Med Chem. 2010;53(24):8650–8662. doi: 10.1021/jm101183p.
    1. Weiler S, et al. Orally active 7-substituted (4-benzylphthalazin-1-yl)-2-methylpiperazin-1-yl)nicotinonitriles as active-site inhibitors of sphingosine 1-phosphate lyase for the treatment of multiple sclerosis. J Med Chem. 2014;57(12):5074–5084. doi: 10.1021/jm500338n.
    1. Richards S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–424. doi: 10.1038/gim.2015.30.
    1. Colié S, et al. Disruption of sphingosine 1-phosphate lyase confers resistance to chemotherapy and promotes oncogenesis through Bcl-2/Bcl-xL upregulation. Cancer Res. 2009;69(24):9346–9353. doi: 10.1158/0008-5472.CAN-09-2198.
    1. Nordgren M, et al. Export-deficient monoubiquitinated PEX5 triggers peroxisome removal in SV40 large T antigen-transformed mouse embryonic fibroblasts. Autophagy. 2015;11(8):1326–1340. doi: 10.1080/15548627.2015.1061846.
    1. Mezzar S, de Schryver E, Van Veldhoven PP. RP-HPLC-fluorescence analysis of aliphatic aldehydes: application to aldehyde-generating enzymes HACL1 and SGPL1. J Lipid Res. 2014;55(3):573–582. doi: 10.1194/jlr.D044230.
    1. Bot M, et al. Hematopoietic sphingosine 1-phosphate lyase deficiency decreases atherosclerotic lesion development in LDL-receptor deficient mice. PLoS One. 2013;8(5):e63360. doi: 10.1371/journal.pone.0063360.
    1. Atkinson D, et al. Sphingosine 1-phosphate lyase deficiency causes Charcot-Marie-Tooth neuropathy [published online ahead of print January 11, 2017] Neurology. doi: .
    1. Sievers F, et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol. 2011;7:539.

Source: PubMed

Подписаться