fbpx
Wikipedia

Sex-determining region Y protein

Sex-determining region Y protein (SRY), or testis-determining factor (TDF), is a DNA-binding protein (also known as gene-regulatory protein/transcription factor) encoded by the SRY gene that is responsible for the initiation of male sex determination in therian mammals (placental mammals and marsupials).[5] SRY is an intronless sex-determining gene on the Y chromosome.[6] Mutations in this gene lead to a range of disorders of sex development with varying effects on an individual's phenotype and genotype.

SRY
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
AliasesSRY, SRXX1, SRXY1, TDF, TDY, Testis determining factor, sex determining region Y, Sex-determining region of Y-chromosome, Sex-determining region Y
External IDsOMIM: 480000 MGI: 98660 HomoloGene: 48168 GeneCards: SRY
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_003140

NM_011564

RefSeq (protein)

NP_003131

NP_035694

Location (UCSC)Chr Y: 2.79 – 2.79 MbChr Y: 2.66 – 2.66 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

In humans, the SRY gene is located on short (p) arm of the Y chromosome at position 11.2

SRY is a member of the SOX (SRY-like box) gene family of DNA-binding proteins. When complexed with the (SF-1) protein, SRY acts as a transcription factor that causes upregulation of other transcription factors, most importantly SOX9.[7] Its expression causes the development of primary sex cords, which later develop into seminiferous tubules. These cords form in the central part of the yet-undifferentiated gonad, turning it into a testis. The now-induced Leydig cells of the testis then start secreting testosterone, while the Sertoli cells produce anti-Müllerian hormone.[8] SRY gene effects normally take place 6–8 weeks after fetus formation which inhibits the female anatomical structural growth in males. It also works towards developing the secondary sexual characteristics of males.

Gene evolution and regulation edit

Evolution edit

SRY may have arisen from a gene duplication of the X chromosome bound gene SOX3, a member of the SOX family.[9][10] This duplication occurred after the split between monotremes and therians. Monotremes lack SRY and some of their sex chromosomes share homology with bird sex chromosomes.[11] SRY is a quickly evolving gene, and its regulation has been difficult to study because sex determination is not a highly conserved phenomenon within the animal kingdom.[12] Even within marsupials and placentals, which use SRY in their sex determination process, the action of SRY differs between species.[10] The gene sequence also changes; while the core of the gene, the high-mobility group (HMG) box, is conserved between species, other regions of the gene are not.[10] SRY is one of only four genes on the human Y chromosome that have been shown to have arisen from the original Y chromosome.[13] The other genes on the human Y chromosome arose from an autosome that fused with the original Y chromosome.[13]

Regulation edit

SRY has little in common with sex determination genes of other model organisms, therefore, mice are the main model research organisms that can be utilized for its study. Understanding its regulation is further complicated because even between mammalian species, there is little protein sequence conservation. The only conserved group in mice and other mammals is the HMG box region that is responsible for DNA binding. Mutations in this region result in sex reversal, where the opposite sex is produced.[14] Because there is little conservation, the SRY promoter, regulatory elements and regulation are not well understood. Within related mammalian groups there are homologies within the first 400–600 base pairs (bp) upstream from the translational start site. In vitro studies of human SRY promoter have shown that a region of at least 310 bp upstream to translational start site are required for SRY promoter function. It has been shown that binding of three transcription factors, steroidogenic factor 1 (SF1), specificity protein 1 (Sp1 transcription factor) and Wilms tumor protein 1 (WT1), to the human promoter sequence, influence expression of SRY.[14]

The promoter region has two Sp1 binding sites, at -150 and -13 that function as regulatory sites. Sp1 is a transcription factor that binds GC-rich consensus sequences, and mutation of the SRY binding sites leads to a 90% reduction in gene transcription. Studies of SF1 have resulted in less definite results. Mutations of SF1 can lead to sex reversal, and deletion can lead to incomplete gonad development. However, it is not clear how SF1 interacts with the SR1 promoter directly.[15] The promoter region also has two WT1 binding sites at -78 and -87 bp from the ATG codon. WT1 is transcription factor that has four C-terminal zinc fingers and an N-terminal Pro/Glu-rich region and primarily functions as an activator. Mutation of the zinc fingers or inactivation of WT1 results in reduced male gonad size. Deletion of the gene resulted in complete sex reversal. It is not clear how WT1 functions to up-regulate SRY, but some research suggests that it helps stabilize message processing.[15] However, there are complications to this hypothesis, because WT1 also is responsible for expression of an antagonist of male development, DAX1, which stands for dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1. An additional copy of DAX1 in mice leads to sex reversal. It is not clear how DAX1 functions, and many different pathways have been suggested, including SRY transcriptional destabilization and RNA binding. There is evidence from work on suppression of male development that DAX1 can interfere with function of SF1, and in turn transcription of SRY by recruiting corepressors.[14]

There is also evidence that GATA binding protein 4 (GATA4) and FOG2 contribute to activation of SRY by associating with its promoter. How these proteins regulate SRY transcription is not clear, but FOG2 and GATA4 mutants have significantly lower levels of SRY transcription.[16] FOGs have zinc finger motifs that can bind DNA, but there is no evidence of FOG2 interaction with SRY. Studies suggest that FOG2 and GATA4 associate with nucleosome remodeling proteins that could lead to its activation.[17]

Function edit

During gestation, the cells of the primordial gonad that lie along the urogenital ridge are in a bipotential state, meaning they possess the ability to become either male cells (Sertoli and Leydig cells) or female cells (follicle cells and theca cells). SRY initiates testis differentiation by activating male-specific transcription factors that allow these bipotential cells to differentiate and proliferate. SRY accomplishes this by upregulating SOX9, a transcription factor with a DNA-binding site very similar to SRY's. SOX9 leads to the upregulation of fibroblast growth factor 9 (Fgf9), which in turn leads to further upregulation of SOX9. Once proper SOX9 levels are reached, the bipotential cells of the gonad begin to differentiate into Sertoli cells. Additionally, cells expressing SRY will continue to proliferate to form the primordial testis. This brief review constitutes the basic series of events, but there are many more factors that influence sex differentiation.

Action in the nucleus edit

The SRY protein consists of three main regions. The central region encompasses the high-mobility group (HMG) domain, which contains nuclear localization sequences and acts as the DNA-binding domain. The C-terminal domain has no conserved structure, and the N-terminal domain can be phosphorylated to enhance DNA-binding.[15] The process begins with nuclear localization of SRY by acetylation of the nuclear localization signal regions, which allows for the binding of importin β and calmodulin to SRY, facilitating its import into the nucleus. Once in the nucleus, SRY and SF1 (steroidogenic factor 1, another transcriptional regulator) complex and bind to TESCO (testis-specific enhancer of Sox9 core), the testes-specific enhancer element of the Sox9 gene in Sertoli cell precursors, located upstream of the Sox9 gene transcription start site.[7] Specifically, it is the HMG region of SRY that binds to the minor groove of the DNA target sequence, causing the DNA to bend and unwind. The establishment of this particular DNA "architecture" facilitates the transcription of the Sox9 gene.[15] In the nucleus of Sertoli cells, SOX9 directly targets the Amh gene as well as the prostaglandin D synthase (Ptgds) gene. SOX9 binding to the enhancer near the Amh promoter allows for the synthesis of Amh while SOX9 binding to the Ptgds gene allows for the production of prostaglandin D2 (PGD2). The reentry of SOX9 into the nucleus is facilitated by autocrine or paracrine signaling conducted by PGD2.[18] SOX9 protein then initiates a positive feedback loop, involving SOX9 acting as its own transcription factor and resulting in the synthesis of large amounts of SOX9.[15]

SOX9 and testes differentiation edit

The SF-1 protein, on its own, leads to minimal transcription of the SOX9 gene in both the XX and XY bipotential gonadal cells along the urogenital ridge. However, binding of the SRY-SF1 complex to the testis-specific enhancer (TESCO) on SOX9 leads to significant up-regulation of the gene in only the XY gonad, while transcription in the XX gonad remains negligible. Part of this up-regulation is accomplished by SOX9 itself through a positive feedback loop; like SRY, SOX9 complexes with SF1 and binds to the TESCO enhancer, leading to further expression of SOX9 in the XY gonad. Two other proteins, FGF9 (fibroblast growth factor 9) and PDG2 (prostaglandin D2), also maintain this up-regulation. Although their exact pathways are not fully understood, they have been proven to be essential for the continued expression of SOX9 at the levels necessary for testes development.[7]

SOX9 and SRY are believed to be responsible for the cell-autonomous differentiation of supporting cell precursors in the gonads into Sertoli cells, the beginning of testes development. These initial Sertoli cells, in the center of the gonad, are hypothesized to be the starting point for a wave of FGF9 that spreads throughout the developing XY gonad, leading to further differentiation of Sertoli cells via the up-regulation of SOX9.[19] SOX9 and SRY are also believed to be responsible for many of the later processes of testis development (such as Leydig cell differentiation, sex cord formation, and formation of testis-specific vasculature), although exact mechanisms remain unclear.[20] It has been shown, however, that SOX9, in the presence of PDG2, acts directly on Amh (encoding anti-Müllerian hormone) and is capable of inducing testis formation in XX mice gonads, indicating it is vital to testes development.[19]

SRY disorders' influence on sex expression edit

Embryos are gonadally identical, regardless of genetic sex, until a certain point in development when the testis-determining factor causes male sex organs to develop. A typical male karyotype is XY, whereas a female's is XX. There are exceptions, however, in which SRY plays a major role. Individuals with Klinefelter syndrome inherit a normal Y chromosome and multiple X chromosomes, giving them a karyotype of XXY. These persons are considered male.[21] Atypical genetic recombination during crossover, when a sperm cell is developing, can result in karyotypes that do not match their phenotypic expression.

Most of the time, when a developing sperm cell undergoes crossover during meiosis, the SRY gene stays on the Y chromosome. If the SRY gene is transferred to the X chromosome instead of staying on the Y chromosome, testis development will no longer occur. This is known as Swyer syndrome, characterized by an XY karyotype and a female phenotype. Individuals who have this syndrome have normally formed uteri and fallopian tubes, but the gonads are not functional. Swyer syndrome individuals are generally raised as females and have a female gender identity.[22] On the other spectrum, XX male syndrome occurs when a body has female chromosomes and SRY attaches to one of them through translocation. People with XX male syndrome have female karyotype but male physical features.[23] Individuals with either of these syndromes can experience delayed puberty, infertility, and growth features of the opposite sex they identify with. XX male syndrome expressers may develop breasts, and those with Swyer syndrome may have facial hair.[22][24]

Klinefelter Syndrome
  • Inherit a normal Y chromosome and multiple X chromosomes, giving persons a karyotype of XXY.
  • Persons with this are considered male.
Swyer Syndrome
  • SRY gene is transferred to the X chromosome instead of staying on the Y chromosome, testis development will no longer occur.
  • Characterized by an XY karyotype and female phenotype.
  • Individuals have normally formed uteri and fallopian tubes, but the gonads are not functional.
XX Male Syndrome
  • Characterized by a body that has female chromosomes and SRY attaches to one of them through translocation.
  • Individuals have female genotype but male physical features.

While the presence or absence of SRY has generally determined whether or not testis development occurs, it has been suggested that there are other factors that affect the functionality of SRY.[25] Therefore, there are individuals who have the SRY gene, but still develop as females, either because the gene itself is defective or mutated, or because one of the contributing factors is defective.[26] This can happen in individuals exhibiting a XY, XXY, or XX SRY-positive karyotype.

Additionally, other sex determining systems that rely on SRY beyond XY are the processes that come after SRY is present or absent in the development of an embryo. In a normal system, if SRY is present for XY, SRY will activate the medulla to develop gonads into testes. Testosterone will then be produced and initiate the development of other male sexual characteristics. Comparably, if SRY is not present for XX, there will be a lack of the SRY based on no Y chromosome. The lack of SRY will allow the cortex of embryonic gonads to develop into ovaries, which will then produce estrogen, and lead to the development of other female sexual characteristics.[27]

Role in other diseases edit

SRY has been shown to interact with the androgen receptor and individuals with XY karyotype and a functional SRY gene can have an outwardly female phenotype due to an underlying androgen insensitivity syndrome (AIS).[28] Individuals with AIS are unable to respond to androgens properly due to a defect in their androgen receptor gene, and affected individuals can have complete or partial AIS.[29] SRY has also been linked to the fact that males are more likely than females to develop dopamine-related diseases such as schizophrenia and Parkinson's disease. SRY encodes a protein that controls the concentration of dopamine, the neurotransmitter that carries signals from the brain that control movement and coordination.[30] Research in mice has shown that a mutation in SOX10, an SRY encoded transcription factor, is linked to the condition of Dominant megacolon in mice.[31] This mouse model is being used to investigate the link between SRY and Hirschsprung disease, or congenital megacolon in humans.[31] There is also a link between SRY encoded transcription factor SOX9 and campomelic dysplasia (CD).[32] This missense mutation causes defective chondrogenesis, or the process of cartilage formation, and manifests as skeletal CD.[33] Two thirds of 46,XY individuals diagnosed with CD have fluctuating amounts of male-to-female sex reversal.[32]

Use in Olympic screening edit

One of the most controversial uses of this discovery was as a means for gender verification at the Olympic Games, under a system implemented by the International Olympic Committee in 1992. Athletes with an SRY gene were not permitted to participate as females, although all athletes in whom this was "detected" at the 1996 Summer Olympics were ruled false positives and were not disqualified. Specifically, eight female participants (out of a total of 3387) at these games were found to have the SRY gene. However, after further investigation of their genetic conditions, all these athletes were verified as female and allowed to compete. These athletes were found to have either partial or full androgen insensitivity, despite having an SRY gene, making them phenotypically female.[34] In the late 1990s, a number of relevant professional societies in United States called for elimination of gender verification, including the American Medical Association, stating that the method used was uncertain and ineffective.[35] Chromosomal screening was eliminated as of the 2000 Summer Olympics,[35][36][37] but this was later followed by other forms of testing based on hormone levels.[38]

Ongoing research edit

Despite the progress made during the past several decades in the study of sex determination, the SRY gene, and its protein, work is still being conducted to further understanding in these areas. There remain factors that need to be identified in the sex-determining molecular network, and the chromosomal changes involved in many other human sex-reversal cases are still unknown. Scientists continue to search for additional sex-determining genes, using techniques such as microarray screening of the genital ridge genes at varying developmental stages, mutagenesis screens in mice for sex-reversal phenotypes, and identifying the genes that transcription factors act on using chromatin immunoprecipitation.[15]

See also edit

References edit

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000184895 - Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000069036 - 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. ^ Berta P, Hawkins JR, Sinclair AH, Taylor A, Griffiths BL, Goodfellow PN, Fellous M (November 1990). "Genetic evidence equating SRY and the testis-determining factor". Nature. 348 (6300): 448–50. Bibcode:1990Natur.348..448B. doi:10.1038/348448A0. PMID 2247149. S2CID 3336314.
  6. ^ Wallis MC, Waters PD, Graves JA (October 2008). "Sex determination in mammals--before and after the evolution of SRY". Cellular and Molecular Life Sciences. 65 (20): 3182–95. doi:10.1007/s00018-008-8109-z. PMID 18581056. S2CID 31675679.
  7. ^ a b c Kashimada K, Koopman P (December 2010). "Sry: the master switch in mammalian sex determination". Development. 137 (23): 3921–30. doi:10.1242/dev.048983. PMID 21062860.
  8. ^ Mittwoch U (October 1988). "The race to be male". New Scientist. 120 (1635): 38–42.
  9. ^ Katoh K, Miyata T (December 1999). "A heuristic approach of maximum likelihood method for inferring phylogenetic tree and an application to the mammalian SOX-3 origin of the testis-determining gene SRY". FEBS Letters. 463 (1–2): 129–32. doi:10.1016/S0014-5793(99)01621-X. PMID 10601652. S2CID 24519808.
  10. ^ a b c Bakloushinskaya, I Y (2009). "Evolution of sex determination in mammals". Biology Bulletin. 36 (2): 167–174. doi:10.1134/S1062359009020095. S2CID 36988324.
  11. ^ Veyrunes F, Waters PD, Miethke P, Rens W, McMillan D, Alsop AE, Grützner F, Deakin JE, Whittington CM, Schatzkamer K, Kremitzki CL, Graves T, Ferguson-Smith MA, Warren W, Marshall Graves JA (June 2008). "Bird-like sex chromosomes of platypus imply recent origin of mammal sex chromosomes". Genome Research. 18 (6): 965–73. doi:10.1101/gr.7101908. PMC 2413164. PMID 18463302.
  12. ^ Bowles J, Schepers G, Koopman P (November 2000). "Phylogeny of the SOX family of developmental transcription factors based on sequence and structural indicators". Developmental Biology. 227 (2): 239–55. doi:10.1006/dbio.2000.9883. PMID 11071752.
  13. ^ a b Graves JA (December 2015). "Weird mammals provide insights into the evolution of mammalian sex chromosomes and dosage compensation". Journal of Genetics. 94 (4): 567–74. doi:10.1007/s12041-015-0572-3. PMID 26690510. S2CID 186238659.
  14. ^ a b c Ely D, Underwood A, Dunphy G, Boehme S, Turner M, Milsted A (November 2010). "Review of the Y chromosome, Sry and hypertension". Steroids. 75 (11): 747–53. doi:10.1016/j.steroids.2009.10.015. PMC 2891862. PMID 19914267.
  15. ^ a b c d e f Harley VR, Clarkson MJ, Argentaro A (August 2003). "The molecular action and regulation of the testis-determining factors, SRY (sex-determining region on the Y chromosome) and SOX9 [SRY-related high-mobility group (HMG) box 9]". Endocrine Reviews. 24 (4): 466–87. doi:10.1210/er.2002-0025. PMID 12920151.
  16. ^ Knower KC, Kelly S, Harley VR (2003). (PDF). Cytogenetic and Genome Research. 101 (3–4): 185–98. doi:10.1159/000074336. PMID 14684982. S2CID 20940513. Archived from the original on 9 August 2017.
  17. ^ Zaytouni T, Efimenko EE, Tevosian SG (2011). "GATA transcription factors in the developing reproductive system". Advances in Genetics. 76: 93–134. doi:10.1016/B978-0-12-386481-9.00004-3. ISBN 9780123864819. PMID 22099693.
  18. ^ Sekido R, Lovell-Badge R (January 2009). "Sex determination and SRY: down to a wink and a nudge?". Trends in Genetics. 25 (1): 19–29. doi:10.1016/j.tig.2008.10.008. PMID 19027189.
  19. ^ a b McClelland K, Bowles J, Koopman P (January 2012). "Male sex determination: insights into molecular mechanisms". Asian Journal of Andrology. 14 (1): 164–71. doi:10.1038/aja.2011.169. PMC 3735148. PMID 22179516.
  20. ^ Sekido R, Lovell-Badge R (2013). "Genetic control of testis development". Sexual Development. 7 (1–3): 21–32. doi:10.1159/000342221. PMID 22964823.
  21. ^ "Klinefelter syndrome". Genetics Home Reference. National Library of Medicine, National Institutes of Health, U.S. Department of Health and Human Services. Retrieved 3 March 2020.
  22. ^ a b "Swyer syndrome". Genetics Home Reference. National Library of Medicine, National Institutes of Health, U.S. Department of Health and Human Services. Retrieved 3 March 2020.
  23. ^ "XX Male Syndrome {". encyclopedia.com. Retrieved 3 March 2020.
  24. ^ "46,XX testicular disorder of sex development". Genetics Home Reference. National Library of Medicine, National Institutes of Health, U.S. Department of Health and Human Services. Retrieved 3 March 2020.
  25. ^ Polanco JC, Koopman P (February 2007). "Sry and the hesitant beginnings of male development". Developmental Biology. 302 (1): 13–24. doi:10.1016/j.ydbio.2006.08.049. PMID 16996051.
  26. ^ Biason-Lauber A, Konrad D, Meyer M, DeBeaufort C, Schoenle EJ (May 2009). "Ovaries and female phenotype in a girl with 46,XY karyotype and mutations in the CBX2 gene". American Journal of Human Genetics. 84 (5): 658–63. doi:10.1016/j.ajhg.2009.03.016. PMC 2680992. PMID 19361780.
  27. ^ Marieb EN, Hoehn K (2018). Human Anatomy & Physiology (Eleventh ed.). [Hoboken, New Jersey]. ISBN 978-0-13-458099-9. OCLC 1004376412.{{cite book}}: CS1 maint: location missing publisher (link)
  28. ^ Yuan X, Lu ML, Li T, Balk SP (December 2001). "SRY interacts with and negatively regulates androgen receptor transcriptional activity". The Journal of Biological Chemistry. 276 (49): 46647–54. doi:10.1074/jbc.M108404200. PMID 11585838.
  29. ^ Lister Hill National Center for Biomedical Communications (2008). "Androgen insensitivity syndrome". Genetics Home Reference. U.S. National Library of Medicine.
  30. ^ Dewing P, Chiang CW, Sinchak K, Sim H, Fernagut PO, Kelly S, Chesselet MF, Micevych PE, Albrecht KH, Harley VR, Vilain E (February 2006). "Direct regulation of adult brain function by the male-specific factor SRY". Current Biology. 16 (4): 415–20. doi:10.1016/j.cub.2006.01.017. PMID 16488877. S2CID 5939578.
  31. ^ a b Herbarth B, Pingault V, Bondurand N, Kuhlbrodt K, Hermans-Borgmeyer I, Puliti A, Wegner M (1998). "Mutation of the Sry-related Sox10 gene in Dominant megacolon, a mouse model for human Hirschsprung disease". Proceedings of the National Academy of Sciences. 95 (9): 5161–5165. Bibcode:1998PNAS...95.5161H. doi:10.1073/pnas.95.9.5161. PMC 20231. PMID 9560246.
  32. ^ a b Pritchett J, Athwal V, Roberts N, Hanley NA, Hanley KP (2011). "Understanding the role of SOX9 in acquired diseases: lessons from development". Trends in Molecular Medicine. 17 (3): 166–174. doi:10.1016/j.molmed.2010.12.001. PMID 21237710.
  33. ^ "OMIM Entry – # 114290 – CAMPOMELIC DYSPLASIA". omim.org. Retrieved 29 February 2020.
  34. ^ "Olympic Gender Testing".
  35. ^ a b Facius GM (1 August 2004). . Gender Testing. facius-homepage.dk. Archived from the original on 26 January 2010. Retrieved 12 June 2011.
  36. ^ Elsas LJ, Ljungqvist A, Ferguson-Smith MA, Simpson JL, Genel M, Carlson AS, Ferris E, de la Chapelle A, Ehrhardt AA (2000). "Gender verification of female athletes". Genetics in Medicine. 2 (4): 249–54. doi:10.1097/00125817-200007000-00008. PMID 11252710.
  37. ^ Dickinson BD, Genel M, Robinowitz CB, Turner PL, Woods GL (October 2002). "Gender verification of female Olympic athletes". Medicine and Science in Sports and Exercise. 34 (10): 1539–42, discussion 1543. doi:10.1097/00005768-200210000-00001. PMID 12370551.
  38. ^ "IOC Regulations on Female Hyperandrogenism" (PDF). International Olympic Committee. 22 June 2012. (PDF) from the original on 13 August 2012. Retrieved 9 August 2012.

Further reading edit

  • Haqq CM, King CY, Ukiyama E, Falsafi S, Haqq TN, Donahoe PK, Weiss MA (December 1994). "Molecular basis of mammalian sexual determination: activation of Müllerian inhibiting substance gene expression by SRY". Science. 266 (5190): 1494–500. Bibcode:1994Sci...266.1494H. doi:10.1126/science.7985018. PMID 7985018.
  • Goodfellow PN, Lovell-Badge R (1993). "SRY and sex determination in mammals". Annual Review of Genetics. 27 (1): 71–92. doi:10.1146/annurev.ge.27.120193.000443. PMID 8122913.
  • Hawkins JR (1993). "Mutational analysis of SRY in XY females". Human Mutation. 2 (5): 347–50. doi:10.1002/humu.1380020504. PMID 8257986. S2CID 43503112.
  • Harley VR (2002). "The Molecular Action of Testis‐Determining Factors SRY and SOX9". The Genetics and Biology of Sex Determination. Novartis Foundation Symposia. Vol. 244. pp. 57–66, discussion 66–7, 79–85, 253–7. doi:10.1002/0470868732.ch6. ISBN 978-0-470-86873-7. PMID 11990798.
  • Jordan BK, Vilain E (2002). "SRY and the Genetics of Sex Determination". Pediatric Gender Assignment. Advances in Experimental Medicine and Biology. Vol. 511. pp. 1–13, discussion 13–4. doi:10.1007/978-1-4615-0621-8_1. ISBN 978-1-4613-5162-7. PMID 12575752.
  • Oh HJ, Lau YF (March 2006). "KRAB: a partner for SRY action on chromatin". Molecular and Cellular Endocrinology. 247 (1–2): 47–52. doi:10.1016/j.mce.2005.12.011. PMID 16414182. S2CID 19870331.
  • Polanco JC, Koopman P (February 2007). "Sry and the hesitant beginnings of male development". Developmental Biology. 302 (1): 13–24. doi:10.1016/j.ydbio.2006.08.049. PMID 16996051.
  • Hawkins JR, Taylor A, Berta P, Levilliers J, Van der Auwera B, Goodfellow PN (February 1992). "Mutational analysis of SRY: nonsense and missense mutations in XY sex reversal". Human Genetics. 88 (4): 471–4. doi:10.1007/BF00215684. PMID 1339396. S2CID 9332496.
  • Hawkins JR, Taylor A, Goodfellow PN, Migeon CJ, Smith KD, Berkovitz GD (November 1992). "Evidence for increased prevalence of SRY mutations in XY females with complete rather than partial gonadal dysgenesis". American Journal of Human Genetics. 51 (5): 979–84. PMC 1682856. PMID 1415266.
  • Ferrari S, Harley VR, Pontiggia A, Goodfellow PN, Lovell-Badge R, Bianchi ME (December 1992). "SRY, like HMG1, recognizes sharp angles in DNA". The EMBO Journal. 11 (12): 4497–506. doi:10.1002/j.1460-2075.1992.tb05551.x. PMC 557025. PMID 1425584.
  • Jäger RJ, Harley VR, Pfeiffer RA, Goodfellow PN, Scherer G (December 1992). "A familial mutation in the testis-determining gene SRY shared by both sexes". Human Genetics. 90 (4): 350–5. doi:10.1007/BF00220457. PMID 1483689. S2CID 19470332.
  • Vilain E, McElreavey K, Jaubert F, Raymond JP, Richaud F, Fellous M (May 1992). "Familial case with sequence variant in the testis-determining region associated with two sex phenotypes". American Journal of Human Genetics. 50 (5): 1008–11. PMC 1682588. PMID 1570829.
  • Müller J, Schwartz M, Skakkebaek NE (July 1992). "Analysis of the sex-determining region of the Y chromosome (SRY) in sex reversed patients: point-mutation in SRY causing sex-reversion in a 46,XY female". The Journal of Clinical Endocrinology and Metabolism. 75 (1): 331–3. doi:10.1210/jcem.75.1.1619028. PMID 1619028.
  • McElreavey KD, Vilain E, Boucekkine C, Vidaud M, Jaubert F, Richaud F, Fellous M (July 1992). "XY sex reversal associated with a nonsense mutation in SRY". Genomics. 13 (3): 838–40. doi:10.1016/0888-7543(92)90164-N. PMID 1639410.
  • Sinclair AH, Berta P, Palmer MS, Hawkins JR, Griffiths BL, Smith MJ, Foster JW, Frischauf AM, Lovell-Badge R, Goodfellow PN (July 1990). "A gene from the human sex-determining region encodes a protein with homology to a conserved DNA-binding motif". Nature. 346 (6281): 240–4. Bibcode:1990Natur.346..240S. doi:10.1038/346240a0. PMID 1695712. S2CID 4364032.
  • Berkovitz GD, Fechner PY, Zacur HW, Rock JA, Snyder HM, Migeon CJ, Perlman EJ (November 1991). "Clinical and pathologic spectrum of 46,XY gonadal dysgenesis: its relevance to the understanding of sex differentiation". Medicine. 70 (6): 375–83. doi:10.1097/00005792-199111000-00003. PMID 1956279. S2CID 37972412.
  • Berta P, Hawkins JR, Sinclair AH, Taylor A, Griffiths BL, Goodfellow PN, Fellous M (November 1990). "Genetic evidence equating SRY and the testis-determining factor". Nature. 348 (6300): 448–50. Bibcode:1990Natur.348..448B. doi:10.1038/348448A0. PMID 2247149. S2CID 3336314.
  • Jäger RJ, Anvret M, Hall K, Scherer G (November 1990). "A human XY female with a frame shift mutation in the candidate testis-determining gene SRY". Nature. 348 (6300): 452–4. Bibcode:1990Natur.348..452J. doi:10.1038/348452a0. PMID 2247151. S2CID 4326539.
  • Ellis NA, Goodfellow PJ, Pym B, Smith M, Palmer M, Frischauf AM, Goodfellow PN (January 1989). "The pseudoautosomal boundary in man is defined by an Alu repeat sequence inserted on the Y chromosome". Nature. 337 (6202): 81–4. Bibcode:1989Natur.337...81E. doi:10.1038/337081a0. PMID 2909893. S2CID 2890077.
  • Whitfield LS, Hawkins TL, Goodfellow PN, Sulston J (May 1995). "41 kilobases of analyzed sequence from the pseudoautosomal and sex-determining regions of the short arm of the human Y chromosome". Genomics. 27 (2): 306–11. doi:10.1006/geno.1995.1047. PMID 7557997.

External links edit

  • GeneReviews/NCBI/NIH/UW entry on 46,XX Testicular Disorder of Sex Development
  • OMIM entries on 46,XX Testicular Disorder of Sex Development
  • Genes,+sry at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
  • Sex-Determining+Region+Y+Protein at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
  • PDBe-KB provides an overview of all the structure information available in the PDB for Human Sex-determining region Y protein

determining, region, protein, redirects, here, other, uses, disambiguation, testis, determining, factor, binding, protein, also, known, gene, regulatory, protein, transcription, factor, encoded, gene, that, responsible, initiation, male, determination, therian. SRY redirects here For other uses see SRY disambiguation Sex determining region Y protein SRY or testis determining factor TDF is a DNA binding protein also known as gene regulatory protein transcription factor encoded by the SRY gene that is responsible for the initiation of male sex determination in therian mammals placental mammals and marsupials 5 SRY is an intronless sex determining gene on the Y chromosome 6 Mutations in this gene lead to a range of disorders of sex development with varying effects on an individual s phenotype and genotype SRYAvailable structuresPDBOrtholog search PDBe RCSBList of PDB id codes1HRY 1HRZ 1J46 1J47 2GZKIdentifiersAliasesSRY SRXX1 SRXY1 TDF TDY Testis determining factor sex determining region Y Sex determining region of Y chromosome Sex determining region YExternal IDsOMIM 480000 MGI 98660 HomoloGene 48168 GeneCards SRYGene location Human Chr Y chromosome human 1 BandYp11 2Start2 786 855 bp 1 End2 787 682 bp 1 Gene location Mouse Chr Y chromosome mouse 2 BandY YpterStart2 662 471 bp 2 End2 663 658 bp 2 RNA expression patternBgeeHumanMouse ortholog Top expressed ingonadskin of abdomenislet of LangerhansAchilles tendonganglionic eminencepharynxthymusplacentasubcutaneous adipose tissuemammary glandTop expressed inankle jointascending aortaaortic valvecumulus cellsupraoptic nucleussalivary glandcondylelacrimal glandmajor salivary glandstria vascularisMore reference expression dataBioGPSMore reference expression dataGene ontologyMolecular functionDNA binding transcription factor activity transcription factor binding calmodulin binding transcription factor activity RNA polymerase II distal enhancer sequence specific binding DNA binding DNA binding transcription factor activity RNA polymerase II specific protein binding DNA binding transcription activator activity RNA polymerase II specificCellular componentcytoplasm nuclear speck nucleoplasm nucleusBiological processcell differentiation regulation of transcription DNA templated sex differentiation transcription DNA templated positive regulation of transcription DNA templated positive regulation of male gonad development male sex determination regulation of transcription by RNA polymerase II negative regulation of transcription by RNA polymerase II positive regulation of transcription by RNA polymerase II central nervous system development positive regulation of gene expression neuron differentiationSources Amigo QuickGOOrthologsSpeciesHumanMouseEntrez673621674EnsemblENSG00000184895ENSMUSG00000069036UniProtQ05066Q05738RefSeq mRNA NM 003140NM 011564RefSeq protein NP 003131NP 035694Location UCSC Chr Y 2 79 2 79 MbChr Y 2 66 2 66 MbPubMed search 3 4 WikidataView Edit HumanView Edit Mouse In humans the SRY gene is located on short p arm of the Y chromosome at position 11 2SRY is a member of the SOX SRY like box gene family of DNA binding proteins When complexed with the SF 1 protein SRY acts as a transcription factor that causes upregulation of other transcription factors most importantly SOX9 7 Its expression causes the development of primary sex cords which later develop into seminiferous tubules These cords form in the central part of the yet undifferentiated gonad turning it into a testis The now induced Leydig cells of the testis then start secreting testosterone while the Sertoli cells produce anti Mullerian hormone 8 SRY gene effects normally take place 6 8 weeks after fetus formation which inhibits the female anatomical structural growth in males It also works towards developing the secondary sexual characteristics of males Contents 1 Gene evolution and regulation 1 1 Evolution 1 2 Regulation 2 Function 2 1 Action in the nucleus 2 2 SOX9 and testes differentiation 3 SRY disorders influence on sex expression 3 1 Role in other diseases 3 2 Use in Olympic screening 3 3 Ongoing research 4 See also 5 References 6 Further reading 7 External linksGene evolution and regulation editEvolution edit SRY may have arisen from a gene duplication of the X chromosome bound gene SOX3 a member of the SOX family 9 10 This duplication occurred after the split between monotremes and therians Monotremes lack SRY and some of their sex chromosomes share homology with bird sex chromosomes 11 SRY is a quickly evolving gene and its regulation has been difficult to study because sex determination is not a highly conserved phenomenon within the animal kingdom 12 Even within marsupials and placentals which use SRY in their sex determination process the action of SRY differs between species 10 The gene sequence also changes while the core of the gene the high mobility group HMG box is conserved between species other regions of the gene are not 10 SRY is one of only four genes on the human Y chromosome that have been shown to have arisen from the original Y chromosome 13 The other genes on the human Y chromosome arose from an autosome that fused with the original Y chromosome 13 Regulation edit SRY has little in common with sex determination genes of other model organisms therefore mice are the main model research organisms that can be utilized for its study Understanding its regulation is further complicated because even between mammalian species there is little protein sequence conservation The only conserved group in mice and other mammals is the HMG box region that is responsible for DNA binding Mutations in this region result in sex reversal where the opposite sex is produced 14 Because there is little conservation the SRY promoter regulatory elements and regulation are not well understood Within related mammalian groups there are homologies within the first 400 600 base pairs bp upstream from the translational start site In vitro studies of human SRY promoter have shown that a region of at least 310 bp upstream to translational start site are required for SRY promoter function It has been shown that binding of three transcription factors steroidogenic factor 1 SF1 specificity protein 1 Sp1 transcription factor and Wilms tumor protein 1 WT1 to the human promoter sequence influence expression of SRY 14 The promoter region has two Sp1 binding sites at 150 and 13 that function as regulatory sites Sp1 is a transcription factor that binds GC rich consensus sequences and mutation of the SRY binding sites leads to a 90 reduction in gene transcription Studies of SF1 have resulted in less definite results Mutations of SF1 can lead to sex reversal and deletion can lead to incomplete gonad development However it is not clear how SF1 interacts with the SR1 promoter directly 15 The promoter region also has two WT1 binding sites at 78 and 87 bp from the ATG codon WT1 is transcription factor that has four C terminal zinc fingers and an N terminal Pro Glu rich region and primarily functions as an activator Mutation of the zinc fingers or inactivation of WT1 results in reduced male gonad size Deletion of the gene resulted in complete sex reversal It is not clear how WT1 functions to up regulate SRY but some research suggests that it helps stabilize message processing 15 However there are complications to this hypothesis because WT1 also is responsible for expression of an antagonist of male development DAX1 which stands for dosage sensitive sex reversal adrenal hypoplasia critical region on chromosome X gene 1 An additional copy of DAX1 in mice leads to sex reversal It is not clear how DAX1 functions and many different pathways have been suggested including SRY transcriptional destabilization and RNA binding There is evidence from work on suppression of male development that DAX1 can interfere with function of SF1 and in turn transcription of SRY by recruiting corepressors 14 There is also evidence that GATA binding protein 4 GATA4 and FOG2 contribute to activation of SRY by associating with its promoter How these proteins regulate SRY transcription is not clear but FOG2 and GATA4 mutants have significantly lower levels of SRY transcription 16 FOGs have zinc finger motifs that can bind DNA but there is no evidence of FOG2 interaction with SRY Studies suggest that FOG2 and GATA4 associate with nucleosome remodeling proteins that could lead to its activation 17 Function editDuring gestation the cells of the primordial gonad that lie along the urogenital ridge are in a bipotential state meaning they possess the ability to become either male cells Sertoli and Leydig cells or female cells follicle cells and theca cells SRY initiates testis differentiation by activating male specific transcription factors that allow these bipotential cells to differentiate and proliferate SRY accomplishes this by upregulating SOX9 a transcription factor with a DNA binding site very similar to SRY s SOX9 leads to the upregulation of fibroblast growth factor 9 Fgf9 which in turn leads to further upregulation of SOX9 Once proper SOX9 levels are reached the bipotential cells of the gonad begin to differentiate into Sertoli cells Additionally cells expressing SRY will continue to proliferate to form the primordial testis This brief review constitutes the basic series of events but there are many more factors that influence sex differentiation Action in the nucleus edit The SRY protein consists of three main regions The central region encompasses the high mobility group HMG domain which contains nuclear localization sequences and acts as the DNA binding domain The C terminal domain has no conserved structure and the N terminal domain can be phosphorylated to enhance DNA binding 15 The process begins with nuclear localization of SRY by acetylation of the nuclear localization signal regions which allows for the binding of importin b and calmodulin to SRY facilitating its import into the nucleus Once in the nucleus SRY and SF1 steroidogenic factor 1 another transcriptional regulator complex and bind to TESCO testis specific enhancer of Sox9 core the testes specific enhancer element of the Sox9 gene in Sertoli cell precursors located upstream of the Sox9 gene transcription start site 7 Specifically it is the HMG region of SRY that binds to the minor groove of the DNA target sequence causing the DNA to bend and unwind The establishment of this particular DNA architecture facilitates the transcription of the Sox9 gene 15 In the nucleus of Sertoli cells SOX9 directly targets the Amh gene as well as the prostaglandin D synthase Ptgds gene SOX9 binding to the enhancer near the Amh promoter allows for the synthesis of Amh while SOX9 binding to the Ptgds gene allows for the production of prostaglandin D2 PGD2 The reentry of SOX9 into the nucleus is facilitated by autocrine or paracrine signaling conducted by PGD2 18 SOX9 protein then initiates a positive feedback loop involving SOX9 acting as its own transcription factor and resulting in the synthesis of large amounts of SOX9 15 SOX9 and testes differentiation edit The SF 1 protein on its own leads to minimal transcription of the SOX9 gene in both the XX and XY bipotential gonadal cells along the urogenital ridge However binding of the SRY SF1 complex to the testis specific enhancer TESCO on SOX9 leads to significant up regulation of the gene in only the XY gonad while transcription in the XX gonad remains negligible Part of this up regulation is accomplished by SOX9 itself through a positive feedback loop like SRY SOX9 complexes with SF1 and binds to the TESCO enhancer leading to further expression of SOX9 in the XY gonad Two other proteins FGF9 fibroblast growth factor 9 and PDG2 prostaglandin D2 also maintain this up regulation Although their exact pathways are not fully understood they have been proven to be essential for the continued expression of SOX9 at the levels necessary for testes development 7 SOX9 and SRY are believed to be responsible for the cell autonomous differentiation of supporting cell precursors in the gonads into Sertoli cells the beginning of testes development These initial Sertoli cells in the center of the gonad are hypothesized to be the starting point for a wave of FGF9 that spreads throughout the developing XY gonad leading to further differentiation of Sertoli cells via the up regulation of SOX9 19 SOX9 and SRY are also believed to be responsible for many of the later processes of testis development such as Leydig cell differentiation sex cord formation and formation of testis specific vasculature although exact mechanisms remain unclear 20 It has been shown however that SOX9 in the presence of PDG2 acts directly on Amh encoding anti Mullerian hormone and is capable of inducing testis formation in XX mice gonads indicating it is vital to testes development 19 SRY disorders influence on sex expression editEmbryos are gonadally identical regardless of genetic sex until a certain point in development when the testis determining factor causes male sex organs to develop A typical male karyotype is XY whereas a female s is XX There are exceptions however in which SRY plays a major role Individuals with Klinefelter syndrome inherit a normal Y chromosome and multiple X chromosomes giving them a karyotype of XXY These persons are considered male 21 Atypical genetic recombination during crossover when a sperm cell is developing can result in karyotypes that do not match their phenotypic expression Most of the time when a developing sperm cell undergoes crossover during meiosis the SRY gene stays on the Y chromosome If the SRY gene is transferred to the X chromosome instead of staying on the Y chromosome testis development will no longer occur This is known as Swyer syndrome characterized by an XY karyotype and a female phenotype Individuals who have this syndrome have normally formed uteri and fallopian tubes but the gonads are not functional Swyer syndrome individuals are generally raised as females and have a female gender identity 22 On the other spectrum XX male syndrome occurs when a body has female chromosomes and SRY attaches to one of them through translocation People with XX male syndrome have female karyotype but male physical features 23 Individuals with either of these syndromes can experience delayed puberty infertility and growth features of the opposite sex they identify with XX male syndrome expressers may develop breasts and those with Swyer syndrome may have facial hair 22 24 Klinefelter Syndrome Inherit a normal Y chromosome and multiple X chromosomes giving persons a karyotype of XXY Persons with this are considered male Swyer Syndrome SRY gene is transferred to the X chromosome instead of staying on the Y chromosome testis development will no longer occur Characterized by an XY karyotype and female phenotype Individuals have normally formed uteri and fallopian tubes but the gonads are not functional XX Male Syndrome Characterized by a body that has female chromosomes and SRY attaches to one of them through translocation Individuals have female genotype but male physical features While the presence or absence of SRY has generally determined whether or not testis development occurs it has been suggested that there are other factors that affect the functionality of SRY 25 Therefore there are individuals who have the SRY gene but still develop as females either because the gene itself is defective or mutated or because one of the contributing factors is defective 26 This can happen in individuals exhibiting a XY XXY or XX SRY positive karyotype Additionally other sex determining systems that rely on SRY beyond XY are the processes that come after SRY is present or absent in the development of an embryo In a normal system if SRY is present for XY SRY will activate the medulla to develop gonads into testes Testosterone will then be produced and initiate the development of other male sexual characteristics Comparably if SRY is not present for XX there will be a lack of the SRY based on no Y chromosome The lack of SRY will allow the cortex of embryonic gonads to develop into ovaries which will then produce estrogen and lead to the development of other female sexual characteristics 27 Role in other diseases edit SRY has been shown to interact with the androgen receptor and individuals with XY karyotype and a functional SRY gene can have an outwardly female phenotype due to an underlying androgen insensitivity syndrome AIS 28 Individuals with AIS are unable to respond to androgens properly due to a defect in their androgen receptor gene and affected individuals can have complete or partial AIS 29 SRY has also been linked to the fact that males are more likely than females to develop dopamine related diseases such as schizophrenia and Parkinson s disease SRY encodes a protein that controls the concentration of dopamine the neurotransmitter that carries signals from the brain that control movement and coordination 30 Research in mice has shown that a mutation in SOX10 an SRY encoded transcription factor is linked to the condition of Dominant megacolon in mice 31 This mouse model is being used to investigate the link between SRY and Hirschsprung disease or congenital megacolon in humans 31 There is also a link between SRY encoded transcription factor SOX9 and campomelic dysplasia CD 32 This missense mutation causes defective chondrogenesis or the process of cartilage formation and manifests as skeletal CD 33 Two thirds of 46 XY individuals diagnosed with CD have fluctuating amounts of male to female sex reversal 32 Use in Olympic screening edit Further information Sex verification in sports One of the most controversial uses of this discovery was as a means for gender verification at the Olympic Games under a system implemented by the International Olympic Committee in 1992 Athletes with an SRY gene were not permitted to participate as females although all athletes in whom this was detected at the 1996 Summer Olympics were ruled false positives and were not disqualified Specifically eight female participants out of a total of 3387 at these games were found to have the SRY gene However after further investigation of their genetic conditions all these athletes were verified as female and allowed to compete These athletes were found to have either partial or full androgen insensitivity despite having an SRY gene making them phenotypically female 34 In the late 1990s a number of relevant professional societies in United States called for elimination of gender verification including the American Medical Association stating that the method used was uncertain and ineffective 35 Chromosomal screening was eliminated as of the 2000 Summer Olympics 35 36 37 but this was later followed by other forms of testing based on hormone levels 38 Ongoing research edit Despite the progress made during the past several decades in the study of sex determination the SRY gene and its protein work is still being conducted to further understanding in these areas There remain factors that need to be identified in the sex determining molecular network and the chromosomal changes involved in many other human sex reversal cases are still unknown Scientists continue to search for additional sex determining genes using techniques such as microarray screening of the genital ridge genes at varying developmental stages mutagenesis screens in mice for sex reversal phenotypes and identifying the genes that transcription factors act on using chromatin immunoprecipitation 15 See also editSex determination systemReferences edit a b c GRCh38 Ensembl release 89 ENSG00000184895 Ensembl May 2017 a b c GRCm38 Ensembl release 89 ENSMUSG00000069036 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 Berta P Hawkins JR Sinclair AH Taylor A Griffiths BL Goodfellow PN Fellous M November 1990 Genetic evidence equating SRY and the testis determining factor Nature 348 6300 448 50 Bibcode 1990Natur 348 448B doi 10 1038 348448A0 PMID 2247149 S2CID 3336314 Wallis MC Waters PD Graves JA October 2008 Sex determination in mammals before and after the evolution of SRY Cellular and Molecular Life Sciences 65 20 3182 95 doi 10 1007 s00018 008 8109 z PMID 18581056 S2CID 31675679 a b c Kashimada K Koopman P December 2010 Sry the master switch in mammalian sex determination Development 137 23 3921 30 doi 10 1242 dev 048983 PMID 21062860 Mittwoch U October 1988 The race to be male New Scientist 120 1635 38 42 Katoh K Miyata T December 1999 A heuristic approach of maximum likelihood method for inferring phylogenetic tree and an application to the mammalian SOX 3 origin of the testis determining gene SRY FEBS Letters 463 1 2 129 32 doi 10 1016 S0014 5793 99 01621 X PMID 10601652 S2CID 24519808 a b c Bakloushinskaya I Y 2009 Evolution of sex determination in mammals Biology Bulletin 36 2 167 174 doi 10 1134 S1062359009020095 S2CID 36988324 Veyrunes F Waters PD Miethke P Rens W McMillan D Alsop AE Grutzner F Deakin JE Whittington CM Schatzkamer K Kremitzki CL Graves T Ferguson Smith MA Warren W Marshall Graves JA June 2008 Bird like sex chromosomes of platypus imply recent origin of mammal sex chromosomes Genome Research 18 6 965 73 doi 10 1101 gr 7101908 PMC 2413164 PMID 18463302 Bowles J Schepers G Koopman P November 2000 Phylogeny of the SOX family of developmental transcription factors based on sequence and structural indicators Developmental Biology 227 2 239 55 doi 10 1006 dbio 2000 9883 PMID 11071752 a b Graves JA December 2015 Weird mammals provide insights into the evolution of mammalian sex chromosomes and dosage compensation Journal of Genetics 94 4 567 74 doi 10 1007 s12041 015 0572 3 PMID 26690510 S2CID 186238659 a b c Ely D Underwood A Dunphy G Boehme S Turner M Milsted A November 2010 Review of the Y chromosome Sry and hypertension Steroids 75 11 747 53 doi 10 1016 j steroids 2009 10 015 PMC 2891862 PMID 19914267 a b c d e f Harley VR Clarkson MJ Argentaro A August 2003 The molecular action and regulation of the testis determining factors SRY sex determining region on the Y chromosome and SOX9 SRY related high mobility group HMG box 9 Endocrine Reviews 24 4 466 87 doi 10 1210 er 2002 0025 PMID 12920151 Knower KC Kelly S Harley VR 2003 Turning on the male SRY SOX9 and sex determination in mammals PDF Cytogenetic and Genome Research 101 3 4 185 98 doi 10 1159 000074336 PMID 14684982 S2CID 20940513 Archived from the original on 9 August 2017 Zaytouni T Efimenko EE Tevosian SG 2011 GATA transcription factors in the developing reproductive system Advances in Genetics 76 93 134 doi 10 1016 B978 0 12 386481 9 00004 3 ISBN 9780123864819 PMID 22099693 Sekido R Lovell Badge R January 2009 Sex determination and SRY down to a wink and a nudge Trends in Genetics 25 1 19 29 doi 10 1016 j tig 2008 10 008 PMID 19027189 a b McClelland K Bowles J Koopman P January 2012 Male sex determination insights into molecular mechanisms Asian Journal of Andrology 14 1 164 71 doi 10 1038 aja 2011 169 PMC 3735148 PMID 22179516 Sekido R Lovell Badge R 2013 Genetic control of testis development Sexual Development 7 1 3 21 32 doi 10 1159 000342221 PMID 22964823 Klinefelter syndrome Genetics Home Reference National Library of Medicine National Institutes of Health U S Department of Health and Human Services Retrieved 3 March 2020 a b Swyer syndrome Genetics Home Reference National Library of Medicine National Institutes of Health U S Department of Health and Human Services Retrieved 3 March 2020 XX Male Syndrome encyclopedia com Retrieved 3 March 2020 46 XX testicular disorder of sex development Genetics Home Reference National Library of Medicine National Institutes of Health U S Department of Health and Human Services Retrieved 3 March 2020 Polanco JC Koopman P February 2007 Sry and the hesitant beginnings of male development Developmental Biology 302 1 13 24 doi 10 1016 j ydbio 2006 08 049 PMID 16996051 Biason Lauber A Konrad D Meyer M DeBeaufort C Schoenle EJ May 2009 Ovaries and female phenotype in a girl with 46 XY karyotype and mutations in the CBX2 gene American Journal of Human Genetics 84 5 658 63 doi 10 1016 j ajhg 2009 03 016 PMC 2680992 PMID 19361780 Marieb EN Hoehn K 2018 Human Anatomy amp Physiology Eleventh ed Hoboken New Jersey ISBN 978 0 13 458099 9 OCLC 1004376412 a href Template Cite book html title Template Cite book cite book a CS1 maint location missing publisher link Yuan X Lu ML Li T Balk SP December 2001 SRY interacts with and negatively regulates androgen receptor transcriptional activity The Journal of Biological Chemistry 276 49 46647 54 doi 10 1074 jbc M108404200 PMID 11585838 Lister Hill National Center for Biomedical Communications 2008 Androgen insensitivity syndrome Genetics Home Reference U S National Library of Medicine Dewing P Chiang CW Sinchak K Sim H Fernagut PO Kelly S Chesselet MF Micevych PE Albrecht KH Harley VR Vilain E February 2006 Direct regulation of adult brain function by the male specific factor SRY Current Biology 16 4 415 20 doi 10 1016 j cub 2006 01 017 PMID 16488877 S2CID 5939578 a b Herbarth B Pingault V Bondurand N Kuhlbrodt K Hermans Borgmeyer I Puliti A Wegner M 1998 Mutation of the Sry related Sox10 gene in Dominant megacolon a mouse model for human Hirschsprung disease Proceedings of the National Academy of Sciences 95 9 5161 5165 Bibcode 1998PNAS 95 5161H doi 10 1073 pnas 95 9 5161 PMC 20231 PMID 9560246 a b Pritchett J Athwal V Roberts N Hanley NA Hanley KP 2011 Understanding the role of SOX9 in acquired diseases lessons from development Trends in Molecular Medicine 17 3 166 174 doi 10 1016 j molmed 2010 12 001 PMID 21237710 OMIM Entry 114290 CAMPOMELIC DYSPLASIA omim org Retrieved 29 February 2020 Olympic Gender Testing a b Facius GM 1 August 2004 The Major Medical Blunder of the 20th Century Gender Testing facius homepage dk Archived from the original on 26 January 2010 Retrieved 12 June 2011 Elsas LJ Ljungqvist A Ferguson Smith MA Simpson JL Genel M Carlson AS Ferris E de la Chapelle A Ehrhardt AA 2000 Gender verification of female athletes Genetics in Medicine 2 4 249 54 doi 10 1097 00125817 200007000 00008 PMID 11252710 Dickinson BD Genel M Robinowitz CB Turner PL Woods GL October 2002 Gender verification of female Olympic athletes Medicine and Science in Sports and Exercise 34 10 1539 42 discussion 1543 doi 10 1097 00005768 200210000 00001 PMID 12370551 IOC Regulations on Female Hyperandrogenism PDF International Olympic Committee 22 June 2012 Archived PDF from the original on 13 August 2012 Retrieved 9 August 2012 Further reading editHaqq CM King CY Ukiyama E Falsafi S Haqq TN Donahoe PK Weiss MA December 1994 Molecular basis of mammalian sexual determination activation of Mullerian inhibiting substance gene expression by SRY Science 266 5190 1494 500 Bibcode 1994Sci 266 1494H doi 10 1126 science 7985018 PMID 7985018 Goodfellow PN Lovell Badge R 1993 SRY and sex determination in mammals Annual Review of Genetics 27 1 71 92 doi 10 1146 annurev ge 27 120193 000443 PMID 8122913 Hawkins JR 1993 Mutational analysis of SRY in XY females Human Mutation 2 5 347 50 doi 10 1002 humu 1380020504 PMID 8257986 S2CID 43503112 Harley VR 2002 The Molecular Action of Testis Determining Factors SRY and SOX9 The Genetics and Biology of Sex Determination Novartis Foundation Symposia Vol 244 pp 57 66 discussion 66 7 79 85 253 7 doi 10 1002 0470868732 ch6 ISBN 978 0 470 86873 7 PMID 11990798 Jordan BK Vilain E 2002 SRY and the Genetics of Sex Determination Pediatric Gender Assignment Advances in Experimental Medicine and Biology Vol 511 pp 1 13 discussion 13 4 doi 10 1007 978 1 4615 0621 8 1 ISBN 978 1 4613 5162 7 PMID 12575752 Oh HJ Lau YF March 2006 KRAB a partner for SRY action on chromatin Molecular and Cellular Endocrinology 247 1 2 47 52 doi 10 1016 j mce 2005 12 011 PMID 16414182 S2CID 19870331 Polanco JC Koopman P February 2007 Sry and the hesitant beginnings of male development Developmental Biology 302 1 13 24 doi 10 1016 j ydbio 2006 08 049 PMID 16996051 Hawkins JR Taylor A Berta P Levilliers J Van der Auwera B Goodfellow PN February 1992 Mutational analysis of SRY nonsense and missense mutations in XY sex reversal Human Genetics 88 4 471 4 doi 10 1007 BF00215684 PMID 1339396 S2CID 9332496 Hawkins JR Taylor A Goodfellow PN Migeon CJ Smith KD Berkovitz GD November 1992 Evidence for increased prevalence of SRY mutations in XY females with complete rather than partial gonadal dysgenesis American Journal of Human Genetics 51 5 979 84 PMC 1682856 PMID 1415266 Ferrari S Harley VR Pontiggia A Goodfellow PN Lovell Badge R Bianchi ME December 1992 SRY like HMG1 recognizes sharp angles in DNA The EMBO Journal 11 12 4497 506 doi 10 1002 j 1460 2075 1992 tb05551 x PMC 557025 PMID 1425584 Jager RJ Harley VR Pfeiffer RA Goodfellow PN Scherer G December 1992 A familial mutation in the testis determining gene SRY shared by both sexes Human Genetics 90 4 350 5 doi 10 1007 BF00220457 PMID 1483689 S2CID 19470332 Vilain E McElreavey K Jaubert F Raymond JP Richaud F Fellous M May 1992 Familial case with sequence variant in the testis determining region associated with two sex phenotypes American Journal of Human Genetics 50 5 1008 11 PMC 1682588 PMID 1570829 Muller J Schwartz M Skakkebaek NE July 1992 Analysis of the sex determining region of the Y chromosome SRY in sex reversed patients point mutation in SRY causing sex reversion in a 46 XY female The Journal of Clinical Endocrinology and Metabolism 75 1 331 3 doi 10 1210 jcem 75 1 1619028 PMID 1619028 McElreavey KD Vilain E Boucekkine C Vidaud M Jaubert F Richaud F Fellous M July 1992 XY sex reversal associated with a nonsense mutation in SRY Genomics 13 3 838 40 doi 10 1016 0888 7543 92 90164 N PMID 1639410 Sinclair AH Berta P Palmer MS Hawkins JR Griffiths BL Smith MJ Foster JW Frischauf AM Lovell Badge R Goodfellow PN July 1990 A gene from the human sex determining region encodes a protein with homology to a conserved DNA binding motif Nature 346 6281 240 4 Bibcode 1990Natur 346 240S doi 10 1038 346240a0 PMID 1695712 S2CID 4364032 Berkovitz GD Fechner PY Zacur HW Rock JA Snyder HM Migeon CJ Perlman EJ November 1991 Clinical and pathologic spectrum of 46 XY gonadal dysgenesis its relevance to the understanding of sex differentiation Medicine 70 6 375 83 doi 10 1097 00005792 199111000 00003 PMID 1956279 S2CID 37972412 Berta P Hawkins JR Sinclair AH Taylor A Griffiths BL Goodfellow PN Fellous M November 1990 Genetic evidence equating SRY and the testis determining factor Nature 348 6300 448 50 Bibcode 1990Natur 348 448B doi 10 1038 348448A0 PMID 2247149 S2CID 3336314 Jager RJ Anvret M Hall K Scherer G November 1990 A human XY female with a frame shift mutation in the candidate testis determining gene SRY Nature 348 6300 452 4 Bibcode 1990Natur 348 452J doi 10 1038 348452a0 PMID 2247151 S2CID 4326539 Ellis NA Goodfellow PJ Pym B Smith M Palmer M Frischauf AM Goodfellow PN January 1989 The pseudoautosomal boundary in man is defined by an Alu repeat sequence inserted on the Y chromosome Nature 337 6202 81 4 Bibcode 1989Natur 337 81E doi 10 1038 337081a0 PMID 2909893 S2CID 2890077 Whitfield LS Hawkins TL Goodfellow PN Sulston J May 1995 41 kilobases of analyzed sequence from the pseudoautosomal and sex determining regions of the short arm of the human Y chromosome Genomics 27 2 306 11 doi 10 1006 geno 1995 1047 PMID 7557997 External links edit nbsp Wikimedia Commons has media related to SRY GeneReviews NCBI NIH UW entry on 46 XX Testicular Disorder of Sex Development OMIM entries on 46 XX Testicular Disorder of Sex Development Genes sry at the U S National Library of Medicine Medical Subject Headings MeSH Sex Determining Region Y Protein at the U S National Library of Medicine Medical Subject Headings MeSH PDBe KB provides an overview of all the structure information available in the PDB for Human Sex determining region Y protein Retrieved from https en wikipedia org w index php title Sex determining region Y protein amp oldid 1201940145, 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.