fbpx
Wikipedia

Cytochrome P450 reductase

Cytochrome P450 reductase (also known as NADPH:ferrihemoprotein oxidoreductase, NADPH:hemoprotein oxidoreductase, NADPH:P450 oxidoreductase, P450 reductase, POR, CPR, CYPOR) is a membrane-bound enzyme required for electron transfer from NADPH to cytochrome P450[5] and other heme proteins including heme oxygenase in the endoplasmic reticulum[6] of the eukaryotic cell.

POR
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesPOR, CPR, CYP450R, cytochrome p450 oxidoreductase, P450 oxidoreductase
External IDsOMIM: 124015 MGI: 97744 HomoloGene: 725 GeneCards: POR
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_000941
NM_001367562

NM_008898

RefSeq (protein)

NP_032924

Location (UCSC)Chr 7: 75.9 – 75.99 MbChr 5: 135.7 – 135.76 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

Gene Edit

Human POR gene has 16 exons and the exons 2-16 code for a 677-amino acid [7] POR protein (NCBI NP_000932.2). There is a single copy of 50 kb POR gene (NCBI NM_000941.2) in humans on chromosome 7 (7q11.23).

Paralogs of POR include nitric oxide synthase (EC 1.14.13.39), NADPH:sulfite reductase (EC 1.8.1.2), and methionine synthase reductase (EC 1.16.1.8).[citation needed]

Protein structure Edit

The 3D crystal structure of human POR has been determined.[8] The molecule is composed of four structural domains: the FMN-binding domain, the connecting domain, the FAD-binding domain, and NADPH-binding domain. The FMN-binding domain is similar to the structure of FMN-containing protein flavodoxin, whereas the FAD-binding domain and NADPH-binding domains are similar to those of flavoprotein ferredoxin-NADP+ reductase (FNR). The connecting domain is situated between the flavodoxin-like and FNR-like domains. Conformation flexibility of POR is a key requirement for interaction with different redox partners like Cytochrome P450 proteins, and biasing the conformation of POR with small molecule ligands may be a way to control interaction with partner proteins and influence metabolism.[9]

Function Edit

In Bacillus megaterium and Bacillus subtilis, POR is a C-terminal domain of CYP102, a single-polypeptide self-sufficient soluble P450 system (P450 is an N-terminal domain). The general scheme of electron flow in the POR/P450 system is:

NADPH → FAD → FMN → P450 → O2

The definitive evidence for the requirement of POR in cytochrome-P450-mediated reactions came from the work of Lu, Junk and Coon,[10] who dissected the P450-containing mixed function oxidase system into three constituent components: POR, cytochrome P450, and lipids.

Since all microsomal P450 enzymes require POR for catalysis, it is expected that disruption of POR would have devastating consequences. POR knockout mice are embryonic lethal,[11] probably due to lack of electron transport to extrahepatic P450 enzymes since liver-specific knockout of POR yields phenotypically and reproductively normal mice that accumulate hepatic lipids and have remarkably diminished capacity of hepatic drug metabolism.[12]

The reduction of cytochrome P450 is not the only physiological function of POR. The final step of heme oxidation by mammalian heme oxygenase requires POR and O2. In yeast, POR affects the ferrireductase activity, probably transferring electrons to the flavocytochrome ferric reductase.[13]

Clinical significance Edit

More than 200 variations in POR gene have been identified.[14][15]

Five missense mutations (A287P, R457H, V492E, C569Y, and V608F) and a splicing mutation in the POR genes have been found in patients who had hormonal evidence for combined deficiencies of two steroidogenic cytochrome P450 enzymes - P450c17 CYP17A1, which catalyzes steroid 17α-hydroxylation and 17,20 lyase reaction, and P450c21 21-hydroxylase, which catalyzes steroid 21-hydroxylation.[16] Another POR missense mutation Y181D has also been identified.[17] Fifteen of nineteen patients having abnormal genitalia and disordered steroidogenesis were homozygous or apparent compound heterozygous for POR mutations that destroyed or dramatically inhibited POR activity.[18]

POR Deficiency – Mixed Oxidase Disease Edit

POR deficiency is the newest form of congenital adrenal hyperplasia first described in 2004.[16] The index patient was a newborn 46,XX Japanese girl with craniosynostosis, hypertelorism, mid-face hypoplasia, radiohumeral synostosis, arachnodactyly and disordered steroidogenesis. However, the clinical and biochemical characteristics of patients with POR deficiency are long known in the literature as so-called mixed oxidase disease, as POR deficiency typically shows a steroid profile that suggests combined deficiencies of steroid 21-hydroxylase and 17α-hydroxylase/17,20 lyase activities. The clinical spectrum of POR deficiency ranges from severely affected children with ambiguous genitalia, adrenal insufficiency, and the Antley-Bixler skeletal malformation syndrome (ABS) to mildly affected individuals with polycystic ovary syndrome-like features. Some of the POR patients were born to mothers who became virilized during pregnancy, suggesting deficient placental aromatization of fetal androgens due to a lesion in microsomal aromatase resulting in low estrogen production, which was later confirmed by lower aromatase activities caused by POR mutations.[19][20] However, it has also been suggested that fetal and maternal virilization in POR deficiency might be caused by increased dihydrotestosterone synthesis by the fetal gonad through an alternative "backdoor pathway" first described in the marsupials and later confirmed in humans.[21] Gas chromatography/mass spectrometry analysis of urinary steroids from pregnant women carrying a POR-deficient fetus described in an earlier report also supports the existence of this pathway,[22][23] and the relevance of the backdoor pathway along with POR dependent steroidogenesis have become clearer from recent studies.[21] The role of POR mutations beyond CAH are being investigated; and questions such as how POR mutations cause bony abnormalities and what role POR variants play in drug metabolism by hepatic P450s are being addressed in recent publications.[24][25][26][27][28] However, reports of ABS in some offspring of mothers who were treated with fluconazole, an antifungal agent which interferes with cholesterol biosynthesis at the level of CYP51 activity - indicate that disordered drug metabolism may result from deficient POR activity.[29]

Williams syndrome Edit

Williams syndrome is a genetic disorder characterized by the deletion of genetic material approximately 1.2 Mb from the POR gene (POR). Cells with this genetic deletion show reduced transcription of POR, it seems, due to the loss of a cis-regulatory element that alters expression of this gene.[30] Some persons with Williams syndrome show characteristics of POR deficiency, including radioulnar synostosis and other skeletal abnormalities.[31] Cases of mild impairment of cortisol and androgen synthesis have been noted,[32] however, despite the fact that deficient POR impairs androgen synthesis, patients with Williams syndrome often show increased androgen levels.[33] A similar increase in testosterone has been observed in a mouse model that has globally decreased POR expression.[34]

See also Edit

References Edit

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000127948 - Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000005514 - Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Pandey AV, Flück CE (May 2013). "NADPH P450 oxidoreductase: structure, function, and pathology of diseases". Pharmacology & Therapeutics. 138 (2): 229–54. doi:10.1016/j.pharmthera.2013.01.010. PMID 23353702.
  6. ^ Jensen K, Møller BL (February 2010). "Plant NADPH-cytochrome P450 oxidoreductases". Phytochemistry. 71 (2–3): 132–41. doi:10.1016/j.phytochem.2009.10.017. PMID 19931102. CPR was shown to be localized in the endoplasmic reticulum in the early 1960s (Williams and Kamin, 1962).
  7. ^ Haniu M, McManus ME, Birkett DJ, Lee TD, Shively JE (October 1989). "Structural and functional analysis of NADPH-cytochrome P-450 reductase from human liver: complete sequence of human enzyme and NADPH-binding sites". Biochemistry. 28 (21): 8639–45. doi:10.1021/bi00447a054. PMID 2513880.
  8. ^ PDB: 3QE2​); Xia C, Panda SP, Marohnic CC, Martásek P, Masters BS, Kim JJ (August 2011). "Structural basis for human NADPH-cytochrome P450 oxidoreductase deficiency". Proceedings of the National Academy of Sciences of the United States of America. 108 (33): 13486–91. Bibcode:2011PNAS..10813486X. doi:10.1073/pnas.1106632108. PMC 3158178. PMID 21808038.
  9. ^ Jensen SB, Thodberg S, Parween S, Moses ME, Hansen CC, Thomsen J, et al. (2021-04-15). "Biased cytochrome P450-mediated metabolism via small-molecule ligands binding P450 oxidoreductase". Nature Communications. 12 (1): 2260. doi:10.1038/s41467-021-22562-w. PMC 8050233. PMID 33859207.
  10. ^ Lu AY, Junk KW, Coon MJ (July 1969). "Resolution of the cytochrome P-450-containing omega-hydroxylation system of liver microsomes into three components". The Journal of Biological Chemistry. 244 (13): 3714–21. doi:10.1016/S0021-9258(18)83427-5. PMID 4389465.
  11. ^ Shen AL, O'Leary KA, Kasper CB (February 2002). "Association of multiple developmental defects and embryonic lethality with loss of microsomal NADPH-cytochrome P450 oxidoreductase". The Journal of Biological Chemistry. 277 (8): 6536–41. doi:10.1074/jbc.M111408200. PMID 11742006.
  12. ^ Gu J, Weng Y, Zhang QY, Cui H, Behr M, Wu L, Yang W, Zhang L, Ding X (July 2003). "Liver-specific deletion of the NADPH-cytochrome P450 reductase gene: impact on plasma cholesterol homeostasis and the function and regulation of microsomal cytochrome P450 and heme oxygenase". The Journal of Biological Chemistry. 278 (28): 25895–901. doi:10.1074/jbc.M303125200. PMID 12697746.
  13. ^ Lesuisse E, Casteras-Simon M, Labbe P (November 1997). "Cytochrome P-450 reductase is responsible for the ferrireductase activity associated with isolated plasma membranes of Saccharomyces cerevisiae". FEMS Microbiology Letters. 156 (1): 147–52. doi:10.1016/S0378-1097(97)00418-7. PMID 9368374.
  14. ^ Pandey AV, Sproll P (214). "Pharmacogenomics of human P450 oxidoreductase". Frontiers in Pharmacology. 5: 103. doi:10.3389/fphar.2014.00103. PMC 4023047. PMID 24847272.
  15. ^ Burkhard FZ, Parween S, Udhane SS, Flück CE, Pandey AV (April 2016). "P450 Oxidoreductase deficiency: Analysis of mutations and polymorphisms". The Journal of Steroid Biochemistry and Molecular Biology. 165 (Pt A): 38–50. doi:10.1016/j.jsbmb.2016.04.003. PMID 27068427.
  16. ^ a b Flück CE, Tajima T, Pandey AV, Arlt W, Okuhara K, Verge CF, Jabs EW, Mendonça BB, Fujieda K, Miller WL (March 2004). "Mutant P450 oxidoreductase causes disordered steroidogenesis with and without Antley-Bixler syndrome". Nature Genetics. 36 (3): 228–30. doi:10.1038/ng1300. PMID 14758361.
  17. ^ Arlt W, Walker EA, Draper N, Ivison HE, Ride JP, Hammer F, Chalder SM, Borucka-Mankiewicz M, Hauffa BP, Malunowicz EM, Stewart PM, Shackleton CH (June 2004). "Congenital adrenal hyperplasia caused by mutant P450 oxidoreductase and human androgen synthesis: analytical study". Lancet. 363 (9427): 2128–35. doi:10.1016/S0140-6736(04)16503-3. PMID 15220035. S2CID 32705841.
  18. ^ Huang N, Pandey AV, Agrawal V, Reardon W, Lapunzina PD, Mowat D, Jabs EW, Van Vliet G, Sack J, Flück CE, Miller WL (May 2005). "Diversity and function of mutations in p450 oxidoreductase in patients with Antley-Bixler syndrome and disordered steroidogenesis". American Journal of Human Genetics. 76 (5): 729–49. doi:10.1086/429417. PMC 1199364. PMID 15793702.
  19. ^ Parween S, Fernández-Cancio M, Benito-Sanz S, Camats N, Rojas Velazquez MN, López-Siguero JP, et al. (April 2020). "Molecular Basis of CYP19A1 Deficiency in a 46,XX Patient With R550W Mutation in POR: Expanding the PORD Phenotype". The Journal of Clinical Endocrinology and Metabolism. 105 (4): e1272–e1290. doi:10.1210/clinem/dgaa076. PMID 32060549.
  20. ^ Pandey AV, Kempná P, Hofer G, Mullis PE, Flück CE (October 2007). "Modulation of human CYP19A1 activity by mutant NADPH P450 oxidoreductase". Molecular Endocrinology. 21 (10): 2579–95. doi:10.1210/me.2007-0245. PMID 17595315.
  21. ^ a b Flück CE, Meyer-Böni M, Pandey AV, Kempná P, Miller WL, Schoenle EJ, Biason-Lauber A (August 2011). "Why boys will be boys: two pathways of fetal testicular androgen biosynthesis are needed for male sexual differentiation". American Journal of Human Genetics. 89 (2): 201–18. doi:10.1016/j.ajhg.2011.06.009. PMC 3155178. PMID 21802064.
  22. ^ Reisch N, Taylor AE, Nogueira EF, Asby DJ, Dhir V, Berry A, Krone N, Auchus RJ, Shackleton CH, Hanley NA, Arlt W (October 2019). "Alternative pathway androgen biosynthesis and human fetal female virilization". Proceedings of the National Academy of Sciences of the United States of America. 116 (44): 22294–22299. doi:10.1073/pnas.1906623116. PMC 6825302. PMID 31611378.
  23. ^ Shackleton C, Marcos J, Arlt W, Hauffa BP (August 2004). "Prenatal diagnosis of P450 oxidoreductase deficiency (ORD): a disorder causing low pregnancy estriol, maternal and fetal virilization, and the Antley-Bixler syndrome phenotype". American Journal of Medical Genetics Part A. 129A (2): 105–12. doi:10.1002/ajmg.a.30171. PMID 15316970. S2CID 22583190.
  24. ^ Flück CE, Mullis PE, Pandey AV (October 2010). "Reduction in hepatic drug metabolizing CYP3A4 activities caused by P450 oxidoreductase mutations identified in patients with disordered steroid metabolism". Biochemical and Biophysical Research Communications. 401 (1): 149–53. doi:10.1016/j.bbrc.2010.09.035. PMID 20849814.
  25. ^ Tomalik-Scharte D, Maiter D, Kirchheiner J, Ivison HE, Fuhr U, Arlt W (December 2010). "Impaired hepatic drug and steroid metabolism in congenital adrenal hyperplasia due to P450 oxidoreductase deficiency". European Journal of Endocrinology. 163 (6): 919–24. doi:10.1530/EJE-10-0764. PMC 2977993. PMID 20844025.
  26. ^ Nicolo C, Flück CE, Mullis PE, Pandey AV (June 2010). "Restoration of mutant cytochrome P450 reductase activity by external flavin". Molecular and Cellular Endocrinology. 321 (2): 245–52. doi:10.1016/j.mce.2010.02.024. PMID 20188793. S2CID 29109570.
  27. ^ Sandee D, Morrissey K, Agrawal V, Tam HK, Kramer MA, Tracy TS, Giacomini KM, Miller WL (November 2010). "Effects of genetic variants of human P450 oxidoreductase on catalysis by CYP2D6 in vitro". Pharmacogenetics and Genomics. 20 (11): 677–86. doi:10.1097/FPC.0b013e32833f4f9b. PMC 5708132. PMID 20940534.
  28. ^ Agrawal V, Choi JH, Giacomini KM, Miller WL (October 2010). "Substrate-specific modulation of CYP3A4 activity by genetic variants of cytochrome P450 oxidoreductase". Pharmacogenetics and Genomics. 20 (10): 611–8. doi:10.1097/FPC.0b013e32833e0cb5. PMC 2940949. PMID 20697309.
  29. ^ Flück CE, Pandey AV (March 2016). "Impact on CYP19A1 activity by mutations in NADPH cytochrome P450 oxidoreductase". The Journal of Steroid Biochemistry and Molecular Biology. 165 (Pt A): 64–70. doi:10.1016/j.jsbmb.2016.03.031. PMID 27032764. S2CID 23498012.
  30. ^ Merla G, Howald C, Henrichsen CN, Lyle R, Wyss C, Zabot MT, Antonarakis SE, Reymond A (August 2006). "Submicroscopic deletion in patients with Williams-Beuren syndrome influences expression levels of the nonhemizygous flanking genes". American Journal of Human Genetics. 79 (2): 332–41. doi:10.1086/506371. PMC 1559497. PMID 16826523.
  31. ^ Charvat KA, Hornstein L, Oestreich AE (1991). "Radio-ulnar synostosis in Williams syndrome. A frequently associated anomaly". Pediatric Radiology. 21 (7): 508–10. doi:10.1007/bf02011725. PMID 1771116. S2CID 33765973.
  32. ^ Ichinose M, Tojo K, Nakamura K, Matsuda H, Tokudome G, Ohta M, Sakai S, Sakai O (June 1996). "Williams syndrome associated with chronic renal failure and various endocrinological abnormalities". Internal Medicine. 35 (6): 482–8. doi:10.2169/internalmedicine.35.482. PMID 8835601.
  33. ^ Partsch CJ, Pankau R, Blum WF, Gosch A, Wessel A (July 1994). "Hormonal regulation in children and adults with Williams-Beuren syndrome". American Journal of Medical Genetics. 51 (3): 251–7. doi:10.1002/ajmg.1320510316. PMID 8074154.
  34. ^ Wu L, Gu J, Cui H, Zhang QY, Behr M, Fang C, Weng Y, Kluetzman K, Swiatek PJ, Yang W, Kaminsky L, Ding X (January 2005). "Transgenic mice with a hypomorphic NADPH-cytochrome P450 reductase gene: effects on development, reproduction, and microsomal cytochrome P450". The Journal of Pharmacology and Experimental Therapeutics. 312 (1): 35–43. doi:10.1124/jpet.104.073353. PMID 15328377. S2CID 8292025.

External links Edit

  • Cytochrome+P450+Reductase at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
  • GeneReviews/NCBI/NIH/UW entry on Cytochrome P450 Oxidoreductase Deficiency

cytochrome, p450, reductase, also, known, nadph, ferrihemoprotein, oxidoreductase, nadph, hemoprotein, oxidoreductase, nadph, p450, oxidoreductase, p450, reductase, cypor, membrane, bound, enzyme, required, electron, transfer, from, nadph, cytochrome, p450, ot. Cytochrome P450 reductase also known as NADPH ferrihemoprotein oxidoreductase NADPH hemoprotein oxidoreductase NADPH P450 oxidoreductase P450 reductase POR CPR CYPOR is a membrane bound enzyme required for electron transfer from NADPH to cytochrome P450 5 and other heme proteins including heme oxygenase in the endoplasmic reticulum 6 of the eukaryotic cell PORAvailable structuresPDBOrtholog search PDBe RCSBList of PDB id codes1B1C 3FJO 3QE2 3QFC 3QFR 3QFS 3QFTIdentifiersAliasesPOR CPR CYP450R cytochrome p450 oxidoreductase P450 oxidoreductaseExternal IDsOMIM 124015 MGI 97744 HomoloGene 725 GeneCards PORGene location Human Chr Chromosome 7 human 1 Band7q11 23Start75 899 200 bp 1 End75 986 855 bp 1 Gene location Mouse Chr Chromosome 5 mouse 2 Band5 G2 5 75 34 cMStart135 698 887 bp 2 End135 764 180 bp 2 RNA expression patternBgeeHumanMouse ortholog Top expressed inright lobe of liverright adrenal glandright lobe of thyroid glandleft lobe of thyroid glandanterior pituitaryright uterine tubeupper lobe of left lungright lungleft uterine tubeislet of LangerhansTop expressed inolfactory epitheliumlacrimal glandright lungright lung lobesuperior surface of tongueesophagusbrown adipose tissuekidneygallbladderleft lobe of liverMore reference expression dataBioGPSMore reference expression dataGene ontologyMolecular functionFMN binding NADPH hemoprotein reductase activity flavin adenine dinucleotide binding cytochrome b5 reductase activity acting on NAD P H protein binding NADP binding electron transfer activity enzyme binding oxidoreductase activity iron cytochrome c reductase activity hydrolase activity nitric oxide dioxygenase activityCellular componentintegral component of membrane membrane intracellular membrane bounded organelle mitochondrion endoplasmic reticulum endoplasmic reticulum membrane cytoplasm cytosolBiological processregulation of growth plate cartilage chondrocyte proliferation nitrate catabolic process negative regulation of cysteine type endopeptidase activity involved in apoptotic process cellular response to follicle stimulating hormone stimulus positive regulation of smoothened signaling pathway response to nutrient demethylation nitric oxide catabolic process negative regulation of apoptotic process positive regulation of monooxygenase activity negative regulation of lipase activity internal peptidyl lysine acetylation regulation of cholesterol metabolic process cellular response to peptide hormone stimulus fatty acid oxidation positive regulation of chondrocyte differentiation cellular organofluorine metabolic process flavonoid metabolic process positive regulation of cholesterol biosynthetic process positive regulation of steroid hormone biosynthetic process cellular response to gonadotropin stimulus carnitine metabolic process xenobiotic metabolic process electron transport chain response to hormoneSources Amigo QuickGOOrthologsSpeciesHumanMouseEntrez544718984EnsemblENSG00000127948ENSMUSG00000005514UniProtP16435P37040RefSeq mRNA NM 000941NM 001367562NM 008898RefSeq protein NP 000932NP 001354491NP 001369584NP 001369586NP 001369587NP 001369588NP 001369591NP 032924Location UCSC Chr 7 75 9 75 99 MbChr 5 135 7 135 76 MbPubMed search 3 4 WikidataView Edit HumanView Edit Mouse Contents 1 Gene 2 Protein structure 3 Function 4 Clinical significance 4 1 POR Deficiency Mixed Oxidase Disease 4 2 Williams syndrome 5 See also 6 References 7 External linksGene EditHuman POR gene has 16 exons and the exons 2 16 code for a 677 amino acid 7 POR protein NCBI NP 000932 2 There is a single copy of 50 kb POR gene NCBI NM 000941 2 in humans on chromosome 7 7q11 23 Paralogs of POR include nitric oxide synthase EC 1 14 13 39 NADPH sulfite reductase EC 1 8 1 2 and methionine synthase reductase EC 1 16 1 8 citation needed Protein structure EditThe 3D crystal structure of human POR has been determined 8 The molecule is composed of four structural domains the FMN binding domain the connecting domain the FAD binding domain and NADPH binding domain The FMN binding domain is similar to the structure of FMN containing protein flavodoxin whereas the FAD binding domain and NADPH binding domains are similar to those of flavoprotein ferredoxin NADP reductase FNR The connecting domain is situated between the flavodoxin like and FNR like domains Conformation flexibility of POR is a key requirement for interaction with different redox partners like Cytochrome P450 proteins and biasing the conformation of POR with small molecule ligands may be a way to control interaction with partner proteins and influence metabolism 9 Function EditIn Bacillus megaterium and Bacillus subtilis POR is a C terminal domain of CYP102 a single polypeptide self sufficient soluble P450 system P450 is an N terminal domain The general scheme of electron flow in the POR P450 system is NADPH FAD FMN P450 O2 The definitive evidence for the requirement of POR in cytochrome P450 mediated reactions came from the work of Lu Junk and Coon 10 who dissected the P450 containing mixed function oxidase system into three constituent components POR cytochrome P450 and lipids Since all microsomal P450 enzymes require POR for catalysis it is expected that disruption of POR would have devastating consequences POR knockout mice are embryonic lethal 11 probably due to lack of electron transport to extrahepatic P450 enzymes since liver specific knockout of POR yields phenotypically and reproductively normal mice that accumulate hepatic lipids and have remarkably diminished capacity of hepatic drug metabolism 12 The reduction of cytochrome P450 is not the only physiological function of POR The final step of heme oxidation by mammalian heme oxygenase requires POR and O2 In yeast POR affects the ferrireductase activity probably transferring electrons to the flavocytochrome ferric reductase 13 Clinical significance EditMore than 200 variations in POR gene have been identified 14 15 Five missense mutations A287P R457H V492E C569Y and V608F and a splicing mutation in the POR genes have been found in patients who had hormonal evidence for combined deficiencies of two steroidogenic cytochrome P450 enzymes P450c17 CYP17A1 which catalyzes steroid 17a hydroxylation and 17 20 lyase reaction and P450c21 21 hydroxylase which catalyzes steroid 21 hydroxylation 16 Another POR missense mutation Y181D has also been identified 17 Fifteen of nineteen patients having abnormal genitalia and disordered steroidogenesis were homozygous or apparent compound heterozygous for POR mutations that destroyed or dramatically inhibited POR activity 18 POR Deficiency Mixed Oxidase Disease Edit See also Cytochrome P450 oxidoreductase deficiency POR deficiency is the newest form of congenital adrenal hyperplasia first described in 2004 16 The index patient was a newborn 46 XX Japanese girl with craniosynostosis hypertelorism mid face hypoplasia radiohumeral synostosis arachnodactyly and disordered steroidogenesis However the clinical and biochemical characteristics of patients with POR deficiency are long known in the literature as so called mixed oxidase disease as POR deficiency typically shows a steroid profile that suggests combined deficiencies of steroid 21 hydroxylase and 17a hydroxylase 17 20 lyase activities The clinical spectrum of POR deficiency ranges from severely affected children with ambiguous genitalia adrenal insufficiency and the Antley Bixler skeletal malformation syndrome ABS to mildly affected individuals with polycystic ovary syndrome like features Some of the POR patients were born to mothers who became virilized during pregnancy suggesting deficient placental aromatization of fetal androgens due to a lesion in microsomal aromatase resulting in low estrogen production which was later confirmed by lower aromatase activities caused by POR mutations 19 20 However it has also been suggested that fetal and maternal virilization in POR deficiency might be caused by increased dihydrotestosterone synthesis by the fetal gonad through an alternative backdoor pathway first described in the marsupials and later confirmed in humans 21 Gas chromatography mass spectrometry analysis of urinary steroids from pregnant women carrying a POR deficient fetus described in an earlier report also supports the existence of this pathway 22 23 and the relevance of the backdoor pathway along with POR dependent steroidogenesis have become clearer from recent studies 21 The role of POR mutations beyond CAH are being investigated and questions such as how POR mutations cause bony abnormalities and what role POR variants play in drug metabolism by hepatic P450s are being addressed in recent publications 24 25 26 27 28 However reports of ABS in some offspring of mothers who were treated with fluconazole an antifungal agent which interferes with cholesterol biosynthesis at the level of CYP51 activity indicate that disordered drug metabolism may result from deficient POR activity 29 Williams syndrome Edit Williams syndrome is a genetic disorder characterized by the deletion of genetic material approximately 1 2 Mb from the POR gene POR Cells with this genetic deletion show reduced transcription of POR it seems due to the loss of a cis regulatory element that alters expression of this gene 30 Some persons with Williams syndrome show characteristics of POR deficiency including radioulnar synostosis and other skeletal abnormalities 31 Cases of mild impairment of cortisol and androgen synthesis have been noted 32 however despite the fact that deficient POR impairs androgen synthesis patients with Williams syndrome often show increased androgen levels 33 A similar increase in testosterone has been observed in a mouse model that has globally decreased POR expression 34 See also EditAndrogen backdoor pathway P450 containing systemsReferences Edit a b c GRCh38 Ensembl release 89 ENSG00000127948 Ensembl May 2017 a b c GRCm38 Ensembl release 89 ENSMUSG00000005514 Ensembl May 2017 Human PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Mouse PubMed Reference National Center for Biotechnology Information U S National Library of Medicine Pandey AV Fluck CE May 2013 NADPH P450 oxidoreductase structure function and pathology of diseases Pharmacology amp Therapeutics 138 2 229 54 doi 10 1016 j pharmthera 2013 01 010 PMID 23353702 Jensen K Moller BL February 2010 Plant NADPH cytochrome P450 oxidoreductases Phytochemistry 71 2 3 132 41 doi 10 1016 j phytochem 2009 10 017 PMID 19931102 CPR was shown to be localized in the endoplasmic reticulum in the early 1960s Williams and Kamin 1962 Haniu M McManus ME Birkett DJ Lee TD Shively JE October 1989 Structural and functional analysis of NADPH cytochrome P 450 reductase from human liver complete sequence of human enzyme and NADPH binding sites Biochemistry 28 21 8639 45 doi 10 1021 bi00447a054 PMID 2513880 PDB 3QE2 Xia C Panda SP Marohnic CC Martasek P Masters BS Kim JJ August 2011 Structural basis for human NADPH cytochrome P450 oxidoreductase deficiency Proceedings of the National Academy of Sciences of the United States of America 108 33 13486 91 Bibcode 2011PNAS 10813486X doi 10 1073 pnas 1106632108 PMC 3158178 PMID 21808038 Jensen SB Thodberg S Parween S Moses ME Hansen CC Thomsen J et al 2021 04 15 Biased cytochrome P450 mediated metabolism via small molecule ligands binding P450 oxidoreductase Nature Communications 12 1 2260 doi 10 1038 s41467 021 22562 w PMC 8050233 PMID 33859207 Lu AY Junk KW Coon MJ July 1969 Resolution of the cytochrome P 450 containing omega hydroxylation system of liver microsomes into three components The Journal of Biological Chemistry 244 13 3714 21 doi 10 1016 S0021 9258 18 83427 5 PMID 4389465 Shen AL O Leary KA Kasper CB February 2002 Association of multiple developmental defects and embryonic lethality with loss of microsomal NADPH cytochrome P450 oxidoreductase The Journal of Biological Chemistry 277 8 6536 41 doi 10 1074 jbc M111408200 PMID 11742006 Gu J Weng Y Zhang QY Cui H Behr M Wu L Yang W Zhang L Ding X July 2003 Liver specific deletion of the NADPH cytochrome P450 reductase gene impact on plasma cholesterol homeostasis and the function and regulation of microsomal cytochrome P450 and heme oxygenase The Journal of Biological Chemistry 278 28 25895 901 doi 10 1074 jbc M303125200 PMID 12697746 Lesuisse E Casteras Simon M Labbe P November 1997 Cytochrome P 450 reductase is responsible for the ferrireductase activity associated with isolated plasma membranes of Saccharomyces cerevisiae FEMS Microbiology Letters 156 1 147 52 doi 10 1016 S0378 1097 97 00418 7 PMID 9368374 Pandey AV Sproll P 214 Pharmacogenomics of human P450 oxidoreductase Frontiers in Pharmacology 5 103 doi 10 3389 fphar 2014 00103 PMC 4023047 PMID 24847272 Burkhard FZ Parween S Udhane SS Fluck CE Pandey AV April 2016 P450 Oxidoreductase deficiency Analysis of mutations and polymorphisms The Journal of Steroid Biochemistry and Molecular Biology 165 Pt A 38 50 doi 10 1016 j jsbmb 2016 04 003 PMID 27068427 a b Fluck CE Tajima T Pandey AV Arlt W Okuhara K Verge CF Jabs EW Mendonca BB Fujieda K Miller WL March 2004 Mutant P450 oxidoreductase causes disordered steroidogenesis with and without Antley Bixler syndrome Nature Genetics 36 3 228 30 doi 10 1038 ng1300 PMID 14758361 Arlt W Walker EA Draper N Ivison HE Ride JP Hammer F Chalder SM Borucka Mankiewicz M Hauffa BP Malunowicz EM Stewart PM Shackleton CH June 2004 Congenital adrenal hyperplasia caused by mutant P450 oxidoreductase and human androgen synthesis analytical study Lancet 363 9427 2128 35 doi 10 1016 S0140 6736 04 16503 3 PMID 15220035 S2CID 32705841 Huang N Pandey AV Agrawal V Reardon W Lapunzina PD Mowat D Jabs EW Van Vliet G Sack J Fluck CE Miller WL May 2005 Diversity and function of mutations in p450 oxidoreductase in patients with Antley Bixler syndrome and disordered steroidogenesis American Journal of Human Genetics 76 5 729 49 doi 10 1086 429417 PMC 1199364 PMID 15793702 Parween S Fernandez Cancio M Benito Sanz S Camats N Rojas Velazquez MN Lopez Siguero JP et al April 2020 Molecular Basis of CYP19A1 Deficiency in a 46 XX Patient With R550W Mutation in POR Expanding the PORD Phenotype The Journal of Clinical Endocrinology and Metabolism 105 4 e1272 e1290 doi 10 1210 clinem dgaa076 PMID 32060549 Pandey AV Kempna P Hofer G Mullis PE Fluck CE October 2007 Modulation of human CYP19A1 activity by mutant NADPH P450 oxidoreductase Molecular Endocrinology 21 10 2579 95 doi 10 1210 me 2007 0245 PMID 17595315 a b Fluck CE Meyer Boni M Pandey AV Kempna P Miller WL Schoenle EJ Biason Lauber A August 2011 Why boys will be boys two pathways of fetal testicular androgen biosynthesis are needed for male sexual differentiation American Journal of Human Genetics 89 2 201 18 doi 10 1016 j ajhg 2011 06 009 PMC 3155178 PMID 21802064 Reisch N Taylor AE Nogueira EF Asby DJ Dhir V Berry A Krone N Auchus RJ Shackleton CH Hanley NA Arlt W October 2019 Alternative pathway androgen biosynthesis and human fetal female virilization Proceedings of the National Academy of Sciences of the United States of America 116 44 22294 22299 doi 10 1073 pnas 1906623116 PMC 6825302 PMID 31611378 Shackleton C Marcos J Arlt W Hauffa BP August 2004 Prenatal diagnosis of P450 oxidoreductase deficiency ORD a disorder causing low pregnancy estriol maternal and fetal virilization and the Antley Bixler syndrome phenotype American Journal of Medical Genetics Part A 129A 2 105 12 doi 10 1002 ajmg a 30171 PMID 15316970 S2CID 22583190 Fluck CE Mullis PE Pandey AV October 2010 Reduction in hepatic drug metabolizing CYP3A4 activities caused by P450 oxidoreductase mutations identified in patients with disordered steroid metabolism Biochemical and Biophysical Research Communications 401 1 149 53 doi 10 1016 j bbrc 2010 09 035 PMID 20849814 Tomalik Scharte D Maiter D Kirchheiner J Ivison HE Fuhr U Arlt W December 2010 Impaired hepatic drug and steroid metabolism in congenital adrenal hyperplasia due to P450 oxidoreductase deficiency European Journal of Endocrinology 163 6 919 24 doi 10 1530 EJE 10 0764 PMC 2977993 PMID 20844025 Nicolo C Fluck CE Mullis PE Pandey AV June 2010 Restoration of mutant cytochrome P450 reductase activity by external flavin Molecular and Cellular Endocrinology 321 2 245 52 doi 10 1016 j mce 2010 02 024 PMID 20188793 S2CID 29109570 Sandee D Morrissey K Agrawal V Tam HK Kramer MA Tracy TS Giacomini KM Miller WL November 2010 Effects of genetic variants of human P450 oxidoreductase on catalysis by CYP2D6 in vitro Pharmacogenetics and Genomics 20 11 677 86 doi 10 1097 FPC 0b013e32833f4f9b PMC 5708132 PMID 20940534 Agrawal V Choi JH Giacomini KM Miller WL October 2010 Substrate specific modulation of CYP3A4 activity by genetic variants of cytochrome P450 oxidoreductase Pharmacogenetics and Genomics 20 10 611 8 doi 10 1097 FPC 0b013e32833e0cb5 PMC 2940949 PMID 20697309 Fluck CE Pandey AV March 2016 Impact on CYP19A1 activity by mutations in NADPH cytochrome P450 oxidoreductase The Journal of Steroid Biochemistry and Molecular Biology 165 Pt A 64 70 doi 10 1016 j jsbmb 2016 03 031 PMID 27032764 S2CID 23498012 Merla G Howald C Henrichsen CN Lyle R Wyss C Zabot MT Antonarakis SE Reymond A August 2006 Submicroscopic deletion in patients with Williams Beuren syndrome influences expression levels of the nonhemizygous flanking genes American Journal of Human Genetics 79 2 332 41 doi 10 1086 506371 PMC 1559497 PMID 16826523 Charvat KA Hornstein L Oestreich AE 1991 Radio ulnar synostosis in Williams syndrome A frequently associated anomaly Pediatric Radiology 21 7 508 10 doi 10 1007 bf02011725 PMID 1771116 S2CID 33765973 Ichinose M Tojo K Nakamura K Matsuda H Tokudome G Ohta M Sakai S Sakai O June 1996 Williams syndrome associated with chronic renal failure and various endocrinological abnormalities Internal Medicine 35 6 482 8 doi 10 2169 internalmedicine 35 482 PMID 8835601 Partsch CJ Pankau R Blum WF Gosch A Wessel A July 1994 Hormonal regulation in children and adults with Williams Beuren syndrome American Journal of Medical Genetics 51 3 251 7 doi 10 1002 ajmg 1320510316 PMID 8074154 Wu L Gu J Cui H Zhang QY Behr M Fang C Weng Y Kluetzman K Swiatek PJ Yang W Kaminsky L Ding X January 2005 Transgenic mice with a hypomorphic NADPH cytochrome P450 reductase gene effects on development reproduction and microsomal cytochrome P450 The Journal of Pharmacology and Experimental Therapeutics 312 1 35 43 doi 10 1124 jpet 104 073353 PMID 15328377 S2CID 8292025 External links EditCytochrome P450 Reductase at the U S National Library of Medicine Medical Subject Headings MeSH GeneReviews NCBI NIH UW entry on Cytochrome P450 Oxidoreductase Deficiency Portal Biology Retrieved from https en wikipedia org w index php title Cytochrome P450 reductase amp oldid 1157875053, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.