1
|
Zhang X, Li J, Chen S, Yang N, Zheng J. Overview of Avian Sex Reversal. Int J Mol Sci 2023; 24:ijms24098284. [PMID: 37175998 PMCID: PMC10179413 DOI: 10.3390/ijms24098284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Sex determination and differentiation are processes by which a bipotential gonad adopts either a testicular or ovarian cell fate, and secondary sexual characteristics adopt either male or female developmental patterns. In birds, although genetic factors control the sex determination program, sex differentiation is sensitive to hormones, which can induce sex reversal when disturbed. Although these sex-reversed birds can form phenotypes opposite to their genotypes, none can experience complete sex reversal or produce offspring under natural conditions. Promising evidence indicates that the incomplete sex reversal is associated with cell autonomous sex identity (CASI) of avian cells, which is controlled by genetic factors. However, studies cannot clearly describe the regulatory mechanism of avian CASI and sex development at present, and these factors require further exploration. In spite of this, the abundant findings of avian sex research have provided theoretical bases for the progress of gender control technologies, which are being improved through interdisciplinary co-operation and will ultimately be employed in poultry production. In this review, we provide an overview of avian sex determination and differentiation and comprehensively summarize the research progress on sex reversal in birds, especially chickens. Importantly, we describe key issues faced by applying gender control systems in poultry production and chronologically summarize the development of avian sex control methods. In conclusion, this review provides unique perspectives for avian sex studies and helps scientists develop more advanced systems for sex regulation in birds.
Collapse
Affiliation(s)
- Xiuan Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Jianbo Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Sirui Chen
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Jiangxia Zheng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| |
Collapse
|
2
|
Zhang X, Li J, Wang X, Jie Y, Sun C, Zheng J, Li J, Yang N, Chen S. ATAC-seq and RNA-seq analysis unravel the mechanism of sex differentiation and infertility in sex reversal chicken. Epigenetics Chromatin 2023; 16:2. [PMID: 36617567 PMCID: PMC9827654 DOI: 10.1186/s13072-022-00476-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/20/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Sex determination and differentiation are complex and delicate processes. In female chickens, the process of sex differentiation is sensitive and prone to be affected by the administration of aromatase inhibitors, which result in chicken sex reversal and infertility. However, the molecular mechanisms underlying sex differentiation and infertility in chicken sex reversal remain unclear. Therefore, we established a sex-reversed chicken flock by injecting an aromatase inhibitor, fadrozole, and constructed relatively high-resolution profiles of the gene expression and chromatin accessibility of embryonic gonads. RESULTS We revealed that fadrozole affected the transcriptional activities of several genes, such as DMRT1, SOX9, FOXL2, and CYP19A1, related to sex determination and differentiation, and the expression of a set of gonadal development-related genes, such as FGFR3 and TOX3, by regulating nearby open chromatin regions in sex-reversed chicken embryos. After sexual maturity, the sex-reversed chickens were confirmed to be infertile, and the possible causes of this infertility were further investigated. We found that the structure of the gonads and sperm were greatly deformed, and we identified several promising genes related to spermatogenesis and infertility, such as SPEF2, DNAI1, and TACR3, through RNA-seq. CONCLUSIONS This study provides clear insights into the exploration of potential molecular basis underlying sex differentiation and infertility in sex-reversed chickens and lays a foundation for further research into the sex development of birds.
Collapse
Affiliation(s)
- Xiuan Zhang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Jianbo Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Xiqiong Wang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Yuchen Jie
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Congjiao Sun
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Jiangxia Zheng
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Junying Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| | - Sirui Chen
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Andersson L, Purugganan M. Molecular genetic variation of animals and plants under domestication. Proc Natl Acad Sci U S A 2022; 119:e2122150119. [PMID: 35858409 PMCID: PMC9335317 DOI: 10.1073/pnas.2122150119] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Domesticated plants and animals played crucial roles as models for evolutionary change by means of natural selection and for establishing the rules of inheritance, originally proposed by Charles Darwin and Gregor Mendel, respectively. Here, we review progress that has been made during the last 35 y in unraveling the molecular genetic variation underlying the stunning phenotypic diversity in crops and domesticated animals that inspired Mendel and Darwin. We notice that numerous domestication genes, crucial for the domestication process, have been identified in plants, whereas animal domestication appears to have a polygenic background with no obvious "domestication genes" involved. Although model organisms, such as Drosophila and Arabidopsis, have replaced domesticated species as models for basic research, the latter are still outstanding models for evolutionary research because phenotypic change in these species represents an evolutionary process over thousands of years. A consequence of this is that some alleles contributing to phenotypic diversity have evolved by accumulating multiple changes in the same gene. The continued molecular characterization of crops and farm animals with ever sharper tools is essential for future food security.
Collapse
Affiliation(s)
- Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23 Uppsala, Sweden
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden
| | - Michael Purugganan
- Center for Genomics and Systems Biology, New York University, New York, NY 10003
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, 129188, United Arab Emirates
| |
Collapse
|
4
|
Sin SYW, Lu L, Edwards SV. De Novo Assembly of the Northern Cardinal ( Cardinalis cardinalis) Genome Reveals Candidate Regulatory Regions for Sexually Dichromatic Red Plumage Coloration. G3 (BETHESDA, MD.) 2020; 10:3541-3548. [PMID: 32792344 PMCID: PMC7534441 DOI: 10.1534/g3.120.401373] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/09/2020] [Indexed: 11/18/2022]
Abstract
Northern cardinals (Cardinalis cardinalis) are common, mid-sized passerines widely distributed in North America. As an iconic species with strong sexual dichromatism, it has been the focus of extensive ecological and evolutionary research, yet genomic studies investigating the evolution of genotype-phenotype association of plumage coloration and dichromatism are lacking. Here we present a new, highly-contiguous assembly for C. cardinalis We generated a 1.1 Gb assembly comprised of 4,762 scaffolds, with a scaffold N50 of 3.6 Mb, a contig N50 of 114.4 kb and a longest scaffold of 19.7 Mb. We identified 93.5% complete and single-copy orthologs from an Aves dataset using BUSCO, demonstrating high completeness of the genome assembly. We annotated the genomic region comprising the CYP2J19 gene, which plays a pivotal role in the red coloration in birds. Comparative analyses demonstrated non-exonic regions unique to the CYP2J19 gene in passerines and a long insertion upstream of the gene in C. cardinalis Transcription factor binding motifs discovered in the unique insertion region in C. cardinalis suggest potential androgen-regulated mechanisms underlying sexual dichromatism. Pairwise Sequential Markovian Coalescent (PSMC) analysis of the genome reveals fluctuations in historic effective population size between 100,000-250,000 in the last 2 millions years, with declines concordant with the beginning of the Pleistocene epoch and Last Glacial Period. This draft genome of C. cardinalis provides an important resource for future studies of ecological, evolutionary, and functional genomics in cardinals and other birds.
Collapse
Affiliation(s)
- Simon Yung Wa Sin
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, 26 Oxford Street, Cambridge, MA 02138
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong
| | - Lily Lu
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, 26 Oxford Street, Cambridge, MA 02138
| | - Scott V Edwards
- Department of Organismic and Evolutionary Biology, Museum of Comparative Zoology, Harvard University, 26 Oxford Street, Cambridge, MA 02138
| |
Collapse
|
5
|
Chen J, Dalirsefat SB, Han D, Dong X, Hua G, Zheng X, Xia T, Shao T, Deng X, Wu C. An EAV-HP insertion in the 5' flanking region of SLCO1B3 is associated with its tissue-expression profile in blue-eggshell Yimeng chickens (Gallus gallus). Poult Sci 2020; 99:6371-6377. [PMID: 33248552 PMCID: PMC7704947 DOI: 10.1016/j.psj.2020.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 08/29/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
We previously reported that blue eggshell color in chickens is associated with a partial endogenous retroviral (EAV-HP) insertion in the promoter region of the solute carrier organic anion transporter family member 1B3 (SLCO1B3) gene. The EAV-HP sequence includes numerous regulatory elements, which may modulate the expression of adjacent genes. To determine whether this insertion influences the expression of neighboring genes, we screened the expression of solute carrier organic anion transporter family members 1C1, 1B1 (SLCO1C1, SLCO1B1), and SLCO1B3 in 13 and 10 tissues from female and male Yimeng chickens, respectively. We observed that the insertion only significantly modulated the expression of SLCO1B3 and did not majorly affect that of SLCO1C1 and SLCO1B1. High expression of SLCO1B3 was detected in the shell gland, magnum, isthmus, and vagina of the oviduct in female blue-eggshell chickens. We also observed ectopic expression of SLCO1B3 in the testes of male chickens. SLCO1B3 is typically highly expressed in the liver; however, the EAV-HP insertion significantly reduces SLCO1B3 expression. As a liver-specific transporter, a reduction in the expression of SLCO1B3 may affect liver metabolism, particularly that of bile acids. We also detected higher ectopic expression of SLCO1B3 in the lungs of birds heterozygous for the EAV-HP insertion than in homozygous genotypes. In conclusion, we confirmed that the EAV-HP insertion modifies SLCO1B3 expression, and showed, for the first time, similar expression profile of this gene in all parts of the oviduct in females and testis in males. We also observed different levels of SLCO1B3 expression in the liver, which were associated with the EAV-HP insertion, and significantly higher expression in the lungs of birds with heterozygous genotype. The effects of these changes in the SLCO1B3 expression pattern on the function of the tissues warrant further investigation.
Collapse
Affiliation(s)
- Jianfei Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| | - Seyed Benyamin Dalirsefat
- Department of Animal Science, Faculty of Agricultural Sciences, University of Guilan, Rasht, Guilan, Iran
| | - Deping Han
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xianggui Dong
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| | - Guoying Hua
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| | - Xiaotong Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| | - Tianlan Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| | - Tianqi Shao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| | - Xuemei Deng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China.
| | - Changxin Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture & Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing 100193, China
| |
Collapse
|
6
|
Li J, Davis BW, Jern P, Dorshorst BJ, Siegel PB, Andersson L. Characterization of the endogenous retrovirus insertion in CYP19A1 associated with henny feathering in chicken. Mob DNA 2019; 10:38. [PMID: 31467598 PMCID: PMC6712707 DOI: 10.1186/s13100-019-0181-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/14/2019] [Indexed: 11/10/2022] Open
Abstract
Background Henny feathering in chickens is determined by a dominant mutation that transforms male-specific plumage to female-like plumage. Previous studies indicated that this phenotype is caused by ectopic expression in skin of CYP19A1 encoding aromatase that converts androgens to estrogen and thereby inhibits the development of male-specific plumage. A long terminal repeat (LTR) from an uncharacterized endogenous retrovirus (ERV) insertion was found in an isoform of the CYP19A1 transcript from henny feathering chicken. However, the complete sequence and the genomic position of the insertion were not determined. Results We used publicly available whole genome sequence data to determine the flanking sequences of the ERV, and then PCR amplified the entire insertion and sequenced it using Nanopore long reads and Sanger sequencing. The 7524 bp insertion contains an intact endogenous retrovirus that was not found in chickens representing 31 different breeds not showing henny feathering or in samples of the ancestral red junglefowl. The sequence shows over 99% sequence identity to the avian leukosis virus ev-1 and ev-21 strains, suggesting a recent integration. The ERV 3’LTR, containing a powerful transcriptional enhancer and core promoter with TATA box together with binding sites for EFIII and Ig/EBP inside the CYP19A1 5′ untranslated region, was detected partially in an aromatase transcript, which present a plausible explanation for ectopic expression of aromatase in non-ovarian tissues underlying the henny feathering phenotype. Conclusions We demonstrate that the henny feathering allele harbors an insertion of an intact avian leukosis virus at the 5’end of CYP19A1. The presence of this ERV showed complete concordance with the henny feathering phenotype both within a pedigree segregating for this phenotype and across breeds. Electronic supplementary material The online version of this article (10.1186/s13100-019-0181-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingyi Li
- 1Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843 USA
| | - Brian W Davis
- 1Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843 USA
| | - Patric Jern
- 2Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Ben J Dorshorst
- 4Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, 24061 USA
| | - Paul B Siegel
- 4Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, 24061 USA
| | - Leif Andersson
- 1Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843 USA.,2Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23 Uppsala, Sweden.,3Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, SE-7507 Uppsala, Sweden
| |
Collapse
|
7
|
Mao H, Wang X, Fan Y, Cheng D, Chen K, Liu S, Xi S, Wan L, Li X, Ren J. Whole-genome SNP data unravel population structure and signatures of selection for black plumage of indigenous chicken breeds from Jiangxi province, China. Anim Genet 2019; 50:475-483. [PMID: 31305959 DOI: 10.1111/age.12827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2019] [Indexed: 01/23/2023]
Abstract
Ten indigenous chicken breeds were originally distributed in Jiangxi Province, China, and they define a critical component of Chinese chicken genetic resources. We have investigated the population genetics of seven Jiangxi chicken breeds using 600K chicken BeadChip SNP data. To provide a genome-wide perspective for the population structure of all 10 Jiangxi chicken breeds, we herein genotyped 78 additional individuals from the seven breeds and 63 chickens from three uninvestigated breeds-Yugan Black (YG), Nancheng Black (NC) and Wanzai Yellow using 55K chicken SNP arrays. We then explored merged data of 17 101 SNPs from 235 individuals to infer the population structure of the 10 breeds. We showed that NC and YG are two regional populations of the same breed, as individuals from the two populations clustered together to form a branch separate from the other breeds in the neighbor-joining tree, they always grouped together in multidimensional principal component analyses and they displayed an identical pattern of ancestral lineage composition. Hence, NC and YG should be considered a single breed in the state-supported conservation scheme. Moreover, we conducted a genome scan for signatures of selection for black plumage. bayescan and hapflk analyses of two contrasting groups (three black-feathered breeds vs. six non-black-feathered breeds) consistently detected 25 putative regions under selection. Nine pigmentation- associated genes (DCT, SLC24A5, SLC30A4, MYO5A, CYP19A1, NADK2, SLC45A2, GNAQ and DCP2) reside within these regions, and these genes are interesting candidates for black plumage and provide a starting point for further identification of causative mutations for black feathers in chicken.
Collapse
Affiliation(s)
- H Mao
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - X Wang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Y Fan
- Department of Animal Science, Jiangxi Biotech Vocational College, Nanchang, 330200, Jiangxi, China
| | - D Cheng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - K Chen
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - S Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - S Xi
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - L Wan
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - X Li
- Unit of Animal Husbandry, Agricultural Bureau of Dongxiang District, Fuzhou, 331800, Jiangxi, China
| | - J Ren
- College of Animal Science, South China Agricultural University, 510642, Guangzhou, China
| |
Collapse
|
8
|
Widelitz RB, Lin GW, Lai YC, Mayer JA, Tang PC, Cheng HC, Jiang TX, Chen CF, Chuong CM. Morpho-regulation in diverse chicken feather formation: Integrating branching modules and sex hormone-dependent morpho-regulatory modules. Dev Growth Differ 2018; 61:124-138. [PMID: 30569461 DOI: 10.1111/dgd.12584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Abstract
Many animals can change the size, shape, texture and color of their regenerated coats in response to different ages, sexes, or seasonal environmental changes. Here, we propose that the feather core branching morphogenesis module can be regulated by sex hormones or other environmental factors to change feather forms, textures or colors, thus generating a large spectrum of complexity for adaptation. We use sexual dimorphisms of the chicken to explore the role of hormones. A long-standing question is whether the sex-dependent feather morphologies are autonomously controlled by the male or female cell types, or extrinsically controlled and reversible. We have recently identified core feather branching molecular modules which control the anterior-posterior (bone morphogenetic orotein [BMP], Wnt gradient), medio-lateral (Retinoic signaling, Gremlin), and proximo-distal (Sprouty, BMP) patterning of feathers. We hypothesize that morpho-regulation, through quantitative modulation of existing parameters, can act on core branching modules to topologically tune the dimension of each parameter during morphogenesis and regeneration. Here, we explore the involvement of hormones in generating sexual dimorphisms using exogenously delivered hormones. Our strategy is to mimic male androgen levels by applying exogenous dihydrotestosterone and aromatase inhibitors to adult females and to mimic female estradiol levels by injecting exogenous estradiol to adult males. We also examine differentially expressed genes in the feathers of wildtype male and female chickens to identify potential downstream modifiers of feather morphogenesis. The data show male and female feather morphology and their color patterns can be modified extrinsically through molting and resetting the stem cell niche during regeneration.
Collapse
Affiliation(s)
- Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Gee-Way Lin
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Chih Lai
- Department of Pathology, University of Southern California, Los Angeles, California.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Julie A Mayer
- Department of Pathology, University of Southern California, Los Angeles, California.,Biocept Inc., San Diego, California
| | - Pin-Chi Tang
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Hsu-Chen Cheng
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Ting-Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Chih-Feng Chen
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
9
|
Morris KR, Hirst CE, Major AT, Ezaz T, Ford M, Bibby S, Doran TJ, Smith CA. Gonadal and Endocrine Analysis of a Gynandromorphic Chicken. Endocrinology 2018; 159:3492-3502. [PMID: 30124802 DOI: 10.1210/en.2018-00553] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/10/2018] [Indexed: 02/08/2023]
Abstract
Birds have a ZZ male and ZW female sex chromosome system. The relative roles of genetics and hormones in regulating avian sexual development have been revealed by studies on gynandromorphs. Gynandromorphs are rare bilateral sex chimeras, male on one side of the body and female on the other. We examined a naturally occurring gynandromorphic chicken that was externally male on the right side of the body and female on the left. The bird was diploid but with a mix of ZZ and ZW cells that correlated with the asymmetric sexual phenotype. The male side was 96% ZZ, and the female side was 77% ZZ and 23% ZW. The gonads of this bird at sexual maturity were largely testicular. The right gonad was a testis, with SOX9+ Sertoli cells, DMRT1+ germ cells, and active spermatogenesis. The left gonad was primarily testicular, but with some peripheral aromatase-expressing follicles. The bird had low levels of serum estradiol and high levels of testosterone, as expected for a male. Despite the low percentage of ZW cells on that side, the left side had female sex-linked feathering, smaller muscle mass, smaller leg and spur, and smaller wattle than the male side. This indicates that these sexually dimorphic structures must be at least partly independent of sex steroid effects. Even a small percentage of ZW cells appears sufficient to support female sexual differentiation. Given the lack of chromosome-wide dosage compensation in birds, various sexually dimorphic features may arise due to Z-gene dosage differences between the sexes.
Collapse
Affiliation(s)
- Kirsten R Morris
- Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Claire E Hirst
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Andrew T Major
- Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Tariq Ezaz
- Institute for Applied Ecology, University of Canberra, Bruce, Australian Capital Territory, Australia
| | - Mark Ford
- Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Susan Bibby
- 2Bridges Consulting, Bendigo, Victoria, Australia
| | - Tim J Doran
- Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
10
|
Chen CC, Plikus MV, Tang PC, Widelitz RB, Chuong CM. The Modulatable Stem Cell Niche: Tissue Interactions during Hair and Feather Follicle Regeneration. J Mol Biol 2016; 428:1423-40. [PMID: 26196442 PMCID: PMC4716892 DOI: 10.1016/j.jmb.2015.07.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/27/2022]
Abstract
Hair and feathers are unique because (1) their stem cells are contained within a follicle structure, (2) they undergo cyclic regeneration repetitively throughout life, (3) regeneration occurs physiologically in healthy individuals and (4) regeneration is also induced in response to injury. Precise control of this cyclic regeneration process is essential for maintaining the homeostasis of living organisms. While stem cells are regulated by the intra-follicle-adjacent micro-environmental niche, this niche is also modulated dynamically by extra-follicular macro-environmental signals, allowing stem cells to adapt to a larger changing environment and physiological needs. Here we review several examples of macro-environments that communicate with the follicles: intradermal adipose tissue, innate immune system, sex hormones, aging, circadian rhythm and seasonal rhythms. Related diseases are also discussed. Unveiling the mechanisms of how stem cell niches are modulated provides clues for regenerative medicine. Given that stem cells are hard to manipulate, focusing translational therapeutic applications at the environments appears to be a more practical approach.
Collapse
Affiliation(s)
- Chih-Chiang Chen
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA; Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan 112; Institute of Clinical Medicine and Department of Dermatology, National Yang-Ming University, Taipei, Taiwan 112
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | - Pin-Chi Tang
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA; Department of Animal Science and Center for the Integrative and Evolutionary, National Chung Hsing University, Taichung, Taiwan 402
| | - Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA
| | - Cheng Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, USA; International Laboratory of Wound Repair and Regeneration, Graduated Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan 701; Integrative Stem Cell Center, China Medical University, Taichung, Taiwan 404.
| |
Collapse
|
11
|
Lambeth LS, Morris KR, Wise TG, Cummins DM, O'Neil TE, Cao Y, Sinclair AH, Doran TJ, Smith CA. Transgenic Chickens Overexpressing Aromatase Have High Estrogen Levels but Maintain a Predominantly Male Phenotype. Endocrinology 2016; 157:83-90. [PMID: 26556534 DOI: 10.1210/en.2015-1697] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Estrogens play a key role in sexual differentiation of both the gonads and external traits in birds. The production of estrogen occurs via a well-characterized steroidogenic pathway, which is a multistep process involving several enzymes, including cytochrome P450 aromatase. In chicken embryos, the aromatase gene (CYP19A1) is expressed female-specifically from the time of gonadal sex differentiation. Ectopic overexpression of aromatase in male chicken embryos induces gonadal sex reversal, and male embryos treated with estradiol become feminized; however, this is not permanent. To test whether a continuous supply of estrogen in adult chickens could induce stable male to female sex reversal, 2 transgenic male chickens overexpressing aromatase were generated using the Tol2/transposase system. These birds had robust ectopic aromatase expression, which resulted in the production of high serum levels of estradiol. Transgenic males had female-like wattle and comb growth and feathering, but they retained male weights, displayed leg spurs, and developed testes. Despite the small sample size, this data strongly suggests that high levels of circulating estrogen are insufficient to maintain a female gonadal phenotype in adult birds. Previous observations of gynandromorph birds and embryos with mixed sex chimeric gonads have highlighted the role of cell autonomous sex identity in chickens. This might imply that in the study described here, direct genetic effects of the male chromosomes largely prevailed over the hormonal profile of the aromatase transgenic birds. This data therefore support the emerging view of at least partial cell autonomous sex development in birds. However, a larger study will confirm this intriguing observation.
Collapse
Affiliation(s)
- Luke S Lambeth
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Kirsten R Morris
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Terry G Wise
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - David M Cummins
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Terri E O'Neil
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Yu Cao
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Andrew H Sinclair
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Timothy J Doran
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| | - Craig A Smith
- Murdoch Childrens Research Institute (L.S.L., A.H.S.), Royal Children's Hospital, Melbourne, Victoria 3052, Australia; Department of Paediatrics (A.H.S.), The University of Melbourne, Melbourne, Victoria 3010, Australia; Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity Flagship (K.R.M., T.G.W., D.M.C., T.E.O., Y.C., T.J.D.), Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; and Department of Anatomy and Developmental Biology (C.A.S.), Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
12
|
Abstract
Endogenous retroviruses comprise millions of discrete genetic loci distributed within the genomes of extant vertebrates. These sequences, which are clearly related to exogenous retroviruses, represent retroviral infections of the deep past, and their abundance suggests that retroviruses were a near-constant presence throughout the evolutionary history of modern vertebrates. Endogenous retroviruses contribute in myriad ways to the evolution of host genomes, as mutagens and as sources of genetic novelty (both coding and regulatory) to be acted upon by the twin engines of random genetic drift and natural selection. Importantly, the richness and complexity of endogenous retrovirus data can be used to understand how viruses spread and adapt on evolutionary timescales by combining population genetics and evolutionary theory with a detailed understanding of retrovirus biology (gleaned from the study of extant retroviruses). In addition to revealing the impact of viruses on organismal evolution, such studies can help us better understand, by looking back in time, how life-history traits, as well as ecological and geological events, influence the movement of viruses within and between populations.
Collapse
Affiliation(s)
- Welkin E Johnson
- Biology Department, Boston College, Chestnut Hill, Massachusetts 02467;
| |
Collapse
|
13
|
Lambeth LS, Ohnesorg T, Cummins DM, Sinclair AH, Smith CA. Development of retroviral vectors for tissue-restricted expression in chicken embryonic gonads. PLoS One 2014; 9:e101811. [PMID: 25003592 PMCID: PMC4086957 DOI: 10.1371/journal.pone.0101811] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 06/11/2014] [Indexed: 01/30/2023] Open
Abstract
The chicken embryo has long been a useful model organism for studying development, including sex determination and gonadal differentiation. However, manipulating gene expression specifically in the embryonic avian gonad has been difficult. The viral vector RCASBP can be readily used for embryo-wide transgene expression; however global mis-expression using this method can cause deleterious off-target effects and embryo-lethality. In an attempt to develop vectors for the over-expression of sequences in chicken embryonic urogenital tissues, the viral vector RCANBP was engineered to contain predicted promoter sequences of gonadal-expressed genes. Several promoters were analysed and it was found that although the SF1 promoter produced a tissue-restricted expression pattern that was highest in the mesonephros and liver, it was also higher in the gonads compared to the rest of the body. The location of EGFP expression from the SF1 promoter overlapped with several key gonad-expressed sex development genes; however expression was generally low-level and was not seen in all gonadal cells. To further validate this sequence the key testis determinant DMRT1 was over-expressed in female embryos, which due to insufficient levels had no effect on gonad development. The female gene aromatase was then over-expressed in male embryos, which disrupted the testis pathway as demonstrated by a reduction in AMH protein. Taken together, although these data showed that the SF1 promoter can be used for functional studies in ovo, a stronger promoter sequence would likely be required for the functional analysis of gonad genes that require high-level expression.
Collapse
Affiliation(s)
- Luke S. Lambeth
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
- Poultry Cooperative Research Centre, Armidale, NSW, Australia
- * E-mail:
| | - Thomas Ohnesorg
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David M. Cummins
- CSIRO Animal, Food and Health Sciences, Australian Animal Health Laboratory, Geelong, VIC, Australia
| | - Andrew H. Sinclair
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Poultry Cooperative Research Centre, Armidale, NSW, Australia
| | - Craig A. Smith
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
- Poultry Cooperative Research Centre, Armidale, NSW, Australia
| |
Collapse
|
14
|
Shozu M, Fukami M, Ogata T. Understanding the pathological manifestations of aromatase excess syndrome: lessons for clinical diagnosis. Expert Rev Endocrinol Metab 2014; 9:397-409. [PMID: 25264451 PMCID: PMC4162655 DOI: 10.1586/17446651.2014.926810] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CYP19A1 Aromatase excess syndrome is characterized by pre- or peripubertal onset of gynecomastia due to estrogen excess because of a gain-of-function mutation in the aromatase gene (CYP19A1). Subchromosomal recombination events including duplication, deletion, and inversion has been identified. The latter two recombinations recruit novel promoters for CYP19A1 through a unique mechanism. Gynecomastia continues for life, and although the general condition is well preserved, it may cause psychological issues. Minor symptoms (variably advanced bone age and short adult height), if present, are exclusively because of estrogen excess. Serum estradiol levels are elevated in 48% of affected males, but are not necessarily useful for diagnosis. Molecular analysis of CYP19A1 mutations is mandatory to confirm aromatase excess syndrome diagnosis. Furthermore, the use of an aromatase inhibitor can ameliorate gynecomastia.
Collapse
Affiliation(s)
- Makio Shozu
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City 260-8670, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Ohkura, Setagaya, Tokyo 157-8535, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| |
Collapse
|
15
|
Nicholas FW, Hobbs M. Mutation discovery for Mendelian traits in non-laboratory animals: a review of achievements up to 2012. Anim Genet 2013; 45:157-70. [PMID: 24372556 PMCID: PMC4225684 DOI: 10.1111/age.12103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2013] [Indexed: 01/21/2023]
Abstract
Within two years of the re-discovery of Mendelism, Bateson and Saunders had described six traits in non-laboratory animals (five in chickens and one in cattle) that show single-locus (Mendelian) inheritance. In the ensuing decades, much progress was made in documenting an ever-increasing number of such traits. In 1987 came the first discovery of a causal mutation for a Mendelian trait in non-laboratory animals: a non-sense mutation in the thyroglobulin gene (TG), causing familial goitre in cattle. In the years that followed, the rate of discovery of causal mutations increased, aided mightily by the creation of genome-wide microsatellite maps in the 1990s and even more mightily by genome assemblies and single-nucleotide polymorphism (SNP) chips in the 2000s. With sequencing costs decreasing rapidly, by 2012 causal mutations were being discovered in non-laboratory animals at a rate of more than one per week. By the end of 2012, the total number of Mendelian traits in non-laboratory animals with known causal mutations had reached 499, which was half the number of published single-locus (Mendelian) traits in those species. The distribution of types of mutations documented in non-laboratory animals is fairly similar to that in humans, with almost half being missense or non-sense mutations. The ratio of missense to non-sense mutations in non-laboratory animals to the end of 2012 was 193:78. The fraction of non-sense mutations (78/271 = 0.29) was not very different from the fraction of non-stop codons that are just one base substitution away from a stop codon (21/61 = 0.34).
Collapse
Affiliation(s)
- Frank W Nicholas
- Faculty of Veterinary Science, University of Sydney, Sydney, NSW, 2006, Australia
| | | |
Collapse
|
16
|
Wragg D, Mwacharo JM, Alcalde JA, Wang C, Han JL, Gongora J, Gourichon D, Tixier-Boichard M, Hanotte O. Endogenous retrovirus EAV-HP linked to blue egg phenotype in Mapuche fowl. PLoS One 2013; 8:e71393. [PMID: 23990950 PMCID: PMC3747184 DOI: 10.1371/journal.pone.0071393] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/02/2013] [Indexed: 12/03/2022] Open
Abstract
Oocyan or blue/green eggshell colour is an autosomal dominant trait found in native chickens (Mapuche fowl) of Chile and in some of their descendants in European and North American modern breeds. We report here the identification of an endogenous avian retroviral (EAV-HP) insertion in oocyan Mapuche fowl and European breeds. Sequencing data reveals 100% retroviral identity between the Mapuche and European insertions. Quantitative real-time PCR analysis of European oocyan chicken indicates over-expression of the SLCO1B3 gene (P<0.05) in the shell gland and oviduct. Predicted transcription factor binding sites in the long terminal repeats (LTR) indicate AhR/Ar, a modulator of oestrogen, as a possible promoter/enhancer leading to reproductive tissue-specific over-expression of the SLCO1B3 gene. Analysis of all jungle fowl species Gallus sp. supports the retroviral insertion to be a post-domestication event, while identical LTR sequences within domestic chickens are in agreement with a recent de novo mutation.
Collapse
Affiliation(s)
- David Wragg
- Centre for Genetics and Genomics, School of Biology, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Joram M. Mwacharo
- Centre for Genetics and Genomics, School of Biology, University of Nottingham, University Park, Nottingham, United Kingdom
| | - José A. Alcalde
- Pontificia Universidad Catolica de Chile, Facultad de Agronomia e Ingenieria Forestal, Santiago, Chile
| | - Chen Wang
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Jian-Lin Han
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- International Livestock Research Institute (ILRI), Nairobi, Kenya
| | - Jaime Gongora
- The University of Sydney, Faculty of Veterinary Science, Sydney, New South Wales, Australia
| | - David Gourichon
- Institut National de la Recherche Agronomique, UE1295 Poultry Experimental Platform of Tours, Nouzilly, France
| | - Michèle Tixier-Boichard
- Institut National de la Recherche Agronomique, AgroParisTech, UMR1313 Animal Genetics and Integrative Biology, Jouy-en-Josas, France
| | - Olivier Hanotte
- Centre for Genetics and Genomics, School of Biology, University of Nottingham, University Park, Nottingham, United Kingdom
| |
Collapse
|
17
|
Lambeth LS, Cummins DM, Doran TJ, Sinclair AH, Smith CA. Overexpression of aromatase alone is sufficient for ovarian development in genetically male chicken embryos. PLoS One 2013; 8:e68362. [PMID: 23840850 PMCID: PMC3695963 DOI: 10.1371/journal.pone.0068362] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/29/2013] [Indexed: 11/25/2022] Open
Abstract
Estrogens play a key role in sexual differentiation of both the gonads and external traits in birds. The production of estrogen occurs via a well-characterised steroidogenic pathway, which is a multi-step process involving several enzymes, including cytochrome P450 aromatase. In chicken embryos, the aromatase gene (CYP19A1) is expressed female-specifically from the time of gonadal sex differentiation. To further explore the role of aromatase in sex determination, we ectopically delivered this enzyme using the retroviral vector RCASBP in ovo. Aromatase overexpression in male chicken embryos induced gonadal sex-reversal characterised by an enlargement of the left gonad and development of ovarian structures such as a thickened outer cortex and medulla with lacunae. In addition, the expression of key male gonad developmental genes (DMRT1, SOX9 and Anti-Müllerian hormone (AMH)) was suppressed, and the distribution of germ cells in sex-reversed males followed the female pattern. The detection of SCP3 protein in late stage sex-reversed male embryonic gonads indicated that these genetically male germ cells had entered meiosis, a process that normally only occurs in female embryonic germ cells. This work shows for the first time that the addition of aromatase into a developing male embryo is sufficient to direct ovarian development, suggesting that male gonads have the complete capacity to develop as ovaries if provided with aromatase.
Collapse
Affiliation(s)
- Luke S Lambeth
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia.
| | | | | | | | | |
Collapse
|
18
|
Matsumoto Y, Buemio A, Chu R, Vafaee M, Crews D. Epigenetic control of gonadal aromatase (cyp19a1) in temperature-dependent sex determination of red-eared slider turtles. PLoS One 2013; 8:e63599. [PMID: 23762231 PMCID: PMC3676416 DOI: 10.1371/journal.pone.0063599] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 04/04/2013] [Indexed: 01/29/2023] Open
Abstract
In the red-eared slider turtle (Trachemys scripta), a species with temperature-dependent sex determination (TSD), the expression of the aromatase gene during gonad development is strictly limited to the female-producing temperature. The underlying mechanism remains unknown. In this study, we identified the upstream 5'-flanking region of the aromatase gene, gonad-specific promoter, and the temperature-dependent DNA methylation signatures during gonad development in the red-eared slider turtle. The 5'-flanking region of the slider aromatase exhibited sequence similarities to the aromatase genes of the American alligator, chicken, quail, and zebra finch. A putative TATA box was located 31 bp upstream of the gonad-specific transcription start site. DNA methylation at the CpG sites between the putative binding sites of the fork head domain factor (FOX) and vertebrate steroidogenic factor 1 (SF1) and adjacent TATA box in the promoter region were significantly lower in embryonic gonads at the female-producing temperature compared the male-producing temperature. A shift from male- to female-, but not from female- to male-, producing temperature changed the level of DNA methylation in gonads. Taken together these results indicate that the temperature, particularly female-producing temperature, allows demethylation at the specific CpG sites of the promoter region which leads the temperature-specific expression of aromatase during gonad development.
Collapse
Affiliation(s)
- Yuiko Matsumoto
- Section of Integrative Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Alvin Buemio
- Section of Integrative Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Randy Chu
- Section of Integrative Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Mozhgon Vafaee
- Section of Integrative Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - David Crews
- Section of Integrative Biology, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
19
|
Wang Z, Qu L, Yao J, Yang X, Li G, Zhang Y, Li J, Wang X, Bai J, Xu G, Deng X, Yang N, Wu C. An EAV-HP insertion in 5' Flanking region of SLCO1B3 causes blue eggshell in the chicken. PLoS Genet 2013; 9:e1003183. [PMID: 23359636 PMCID: PMC3554524 DOI: 10.1371/journal.pgen.1003183] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 10/28/2012] [Indexed: 12/04/2022] Open
Abstract
The genetic determination of eggshell coloration has not been determined in birds. Here we report that the blue eggshell is caused by an EAV-HP insertion that promotes the expression of SLCO1B3 gene in the uterus (shell gland) of the oviduct in chicken. In this study, the genetic map location of the blue eggshell gene was refined by linkage analysis in an F2 chicken population, and four candidate genes within the refined interval were subsequently tested for their expression levels in the shell gland of the uterus from blue-shelled and non-blue-shelled hens. SLCO1B3 gene was found to be the only one expressed in the uterus of blue-shelled hens but not in that of non-blue-shelled hens. Results from a pyrosequencing analysis showed that only the allele of SLCO1B3 from blue-shelled chickens was expressed in the uterus of heterozygous hens (O*LC/O*N). SLCO1B3 gene belongs to the organic anion transporting polypeptide (OATP) family; and the OATPs, functioning as membrane transporters, have been reported for the transportation of amphipathic organic compounds, including bile salt in mammals. We subsequently resequenced the whole genomic region of SLCO1B3 and discovered an EAV-HP insertion in the 5′ flanking region of SLCO1B3. The EAV-HP insertion was found closely associated with blue eggshell phenotype following complete Mendelian segregation. In situ hybridization also demonstrated that the blue eggshell is associated with ectopic expression of SLCO1B3 in shell glands of uterus. Our finding strongly suggests that the EAV-HP insertion is the causative mutation for the blue eggshell phenotype. The insertion was also found in another Chinese blue-shelled breed and an American blue-shelled breed. In addition, we found that the insertion site in the blue-shelled chickens from Araucana is different from that in Chinese breeds, which implied independent integration events in the blue-shelled chickens from the two continents, providing a parallel evolutionary example at the molecular level. The eggshell color of birds is of wide interest, but the molecular basis remained unknown until our discovery, reported here. The blue eggshell is found not only in wild birds but also in domestic fowls. In this study, we identified that blue eggshell in chickens from different geographical regions is caused by a ∼4.2 kb EAV-HP insertion in the 5′ flanking region of SLCO1B3. The EAV-HP insertion in chicken is a derived mutation in domestic chickens. The genetic determination of blue eggshell in other birds requires further investigation. We also found that the EAV-HP insertions in the chickens from China and America were separate integration events, which presents us with a parallel molecular evolution example driven by artificial selection.
Collapse
Affiliation(s)
- Zhepeng Wang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Lujiang Qu
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Junfeng Yao
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Xiaolin Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Guangqi Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Yuanyuan Zhang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Junying Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Xiaotong Wang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Jirong Bai
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Guiyun Xu
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| | - Xuemei Deng
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
- * E-mail: (XD); (NY)
| | - Ning Yang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
- * E-mail: (XD); (NY)
| | - Changxin Wu
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Ellis HL, Shioda K, Rosenthal NF, Coser KR, Shioda T. Masculine epigenetic sex marks of the CYP19A1/aromatase promoter in genetically male chicken embryonic gonads are resistant to estrogen-induced phenotypic sex conversion. Biol Reprod 2012; 87:23, 1-12. [PMID: 22539680 DOI: 10.1095/biolreprod.112.099747] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Sex of birds is genetically determined through inheritance of the ZW sex chromosomes (ZZ males and ZW females). Although the mechanisms of avian sex determination remains unknown, the genetic sex is experimentally reversible by in ovo exposure to exogenous estrogens (ZZ-male feminization) or aromatase inhibitors (ZW-female masculinization). Expression of various testis- and ovary-specific marker genes during the normal and reversed gonadal sex differentiation in chicken embryos has been extensively studied, but the roles of sex-specific epigenetic marks in sex differentiation are unknown. In this study, we show that a 170-nt region in the promoter of CYP19A1/aromatase, a key gene required for ovarian estrogen biosynthesis and feminization of chicken embryonic gonads, contains highly quantitative, nucleotide base-level epigenetic marks that reflect phenotypic gonadal sex differentiation. We developed a protocol to feminize ZZ-male chicken embryonic gonads in a highly quantitative manner by direct injection of emulsified ethynylestradiol into yolk at various developmental stages. Taking advantage of this experimental sex reversal model, we show that the epigenetic sex marks in the CYP19A1/aromatase promoter involving DNA methylation and histone lysine methylation are feminized significantly but only partially in sex-converted gonads even when morphological and transcriptional marks of sex differentiation show complete feminization, being indistinguishable from gonads of normal ZW females. Our study suggests that the epigenetic sex of chicken embryonic gonads is more stable than the morphologically or transcriptionally characterized sex differentiation, suggesting the importance of the nucleotide base-level epigenetic sex in gonadal sex differentiation.
Collapse
Affiliation(s)
- Haley L Ellis
- Molecular Profiling Laboratory, Massachusetts General Hospital Center for Cancer Research and Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | |
Collapse
|
21
|
Chang CM, Coville JL, Coquerelle G, Gourichon D, Oulmouden A, Tixier-Boichard M. Complete association between a retroviral insertion in the tyrosinase gene and the recessive white mutation in chickens. BMC Genomics 2006; 7:19. [PMID: 16457736 PMCID: PMC1373650 DOI: 10.1186/1471-2164-7-19] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Accepted: 02/05/2006] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND In chickens, three mutant alleles have been reported at the C locus, including the albino mutation, and the recessive white mutation, which is characterized by white plumage and pigmented eyes. The albino mutation was found to be a 6 bp deletion in the tyrosinase (TYR) gene. The present work describes an approach to identify the structural rearrangement in the TYR gene associated with the recessive white mutation. RESULTS Molecular analysis of the chicken TYR gene has revealed a major structural difference (Restriction Fragment Length Polymorphism, RFLP) in the genomic DNA of the recessive white chicken. A major size difference of 7.7 kb was found in intron 4 of the TYR gene by long-range PCR. Molecular cloning and sequencing results showed the insertion of a complete avian retroviral sequence of the Avian Leukosis Virus (ALV) family. Several aberrant transcripts of the tyrosinase gene were found in 10 week old recessive white chickens but not in the homozygous wild type colored chicken. We established a rapid genotyping diagnostic test based on the discovery of this retroviral insertion. It shows that all homozygous carriers of this insertion had a white plumage in various chicken strains. Furthermore, it was possible to distinguish heterozygous carriers from homozygous normal chickens in a segregating line. CONCLUSION In this study, we conclude that the insertion of a complete avian retroviral sequence in intron 4 of the tyrosinase gene is diagnostic of the recessive white mutation in chickens. This insertion causes aberrant transcripts lacking exon 5, and we propose that this insertion is the causal mutation for the recessive white allele in the chicken.
Collapse
Affiliation(s)
- Chung-Ming Chang
- UMR Génétique et Diversité Animales, INRA/INA P-G, Centre de Recherches de Jouy, 78352 Jouy-en-Josas, France
| | - Jean-Luc Coville
- UMR Génétique et Diversité Animales, INRA/INA P-G, Centre de Recherches de Jouy, 78352 Jouy-en-Josas, France
| | - Gérard Coquerelle
- UMR Génétique et Diversité Animales, INRA/INA P-G, Centre de Recherches de Jouy, 78352 Jouy-en-Josas, France
| | - David Gourichon
- Unité Expérimentale de Génétique Factorielle Avicole, INRA, Centre de Recherches de Tours, 37380 Nouzilly, France
| | - Ahmad Oulmouden
- UMR Génétique Moléculaire Animale, INRA/Université de Limoges, 87061 Limoges, France
| | - Michèle Tixier-Boichard
- UMR Génétique et Diversité Animales, INRA/INA P-G, Centre de Recherches de Jouy, 78352 Jouy-en-Josas, France
| |
Collapse
|
22
|
Abstract
Oestrogens derived from the neural aromatisation of testosterone play a key role in the activation of male sexual behaviour in many vertebrates. Besides their slow action on gene transcription mediated by the binding to nuclear receptors, oestrogens have now been recognised to have more rapid membrane-based effects on brain function. Rapid changes in aromatase activity, and hence in local oestrogen concentrations, could thus rapidly modulate behavioural responses. We previously demonstrated that calcium-dependent kinases are able to down-regulate aromatase activity after incubations of 10-15 min in phosphorylating conditions. In the present study, in quail hypothalamic homogenates, we show that Ca2+ or calmodulin alone can very rapidly change aromatase activity. Preincubation with 1 mM EGTA or with a monoclonal antibody raised against calmodulin immediately increased aromatase activity. The presence of calmodulin on aromatase purified by immunoprecipitation and electrophoresis was previously identified by western blot and two consensus binding sites for Ca2+-calmodulin are identified here on the deduced amino acid sequence of the quail brain aromatase. The rapid control of brain aromatase activity thus appears to include two mechanisms: (i) an immediate regulatory process that involves the Ca2+-calmodulin binding site and (ii) a somewhat slower phosphorylation by several protein kinases (PKC, PKA but also possibly Ca2+-calmodulin kinases) of the aromatase molecule.
Collapse
Affiliation(s)
- J Balthazart
- Center for Cellular and Molecular Neurobiology, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium.
| | | | | | | |
Collapse
|
23
|
Mayer JA, Chuong CM, Widelitz R. Rooster feathering, androgenic alopecia, and hormone-dependent tumor growth: what is in common? Differentiation 2004; 72:474-88. [PMID: 15617560 PMCID: PMC4380229 DOI: 10.1111/j.1432-0436.2004.07209003.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Different epithelial organs form as a result of epithelial-mesenchymal interactions and share a common theme modulated by variations (Chuong ed. In Molecular Basis of Epithelial Appendage Morphogenesis, 1998). One of the major modulators is the sex hormone pathway that acts on the prototype signaling pathway to alter organ phenotypes. Here, we focus on how the sex hormone pathway may interface with epithelia morphogenesis-related signaling pathways. We first survey these sex hormone-regulated morphogenetic processes in various epithelial organs. Sexual dimorphism of hairs and feathers has implications in sexual selection. Diseases of these pathways result in androgenic alopecia, hirsutism, henny feathering, etc. The growth and development of mammary glands, prostate glands, and external genitalia essential for reproductive function are also dependent on sex hormones. Diseases affecting these organs include congenital anomalies and hormone-dependent breast and prostate cancers. To study the role of sex hormones in new growth in the context of system biology/pathology, an in vivo model in which organ formation starts from stem cells is essential. With recent developments (Yu et al. (2002) The morphogenesis of feathers. Nature 420:308-312), the growth of tail feathers in roosters and hens has become a testable model in which experimental manipulations are possible. We show exemplary data of differences in their growth rate, proliferative cell population, and signaling molecule expression. Working hypotheses are proposed on how the sex hormone pathways may interact with growth pathways. It is now possible to test these hypotheses using the chicken model to learn fundamental mechanisms on how sex hormones affect organogenesis, epithelial organ cycling, and growth-related tumorigenesis.
Collapse
Affiliation(s)
- Julie Ann Mayer
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Randall Widelitz
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
24
|
Balthazart J, Baillien M, Charlier TD, Cornil CA, Ball GF. Multiple mechanisms control brain aromatase activity at the genomic and non-genomic level. J Steroid Biochem Mol Biol 2003; 86:367-79. [PMID: 14623533 DOI: 10.1016/s0960-0760(03)00346-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Evidence has recently accumulated indicating that aromatase activity in the preoptic area is modulated in parallel by both slow (hours to days) genomic and rapid (minutes to hours) non-genomic mechanisms. We review here these two types of control mechanisms and their potential contribution to various aspects of brain physiology in quail. High levels of aromatase mRNA, protein and activity (AA) are present in the preoptic area of this species where the transcription of aromatase is controlled mainly by steroids. Estrogens acting in synergy with androgens play a key role in this control and both androgen and estrogen receptors (ER; alpha and beta subtypes) are present in the preoptic area even if they are not necessarily co-localized in the same cells as aromatase. Steroids have more pronounced effects on aromatase transcription in males than in females and this sex difference could be caused, in part, by a sexually differentiated expression of the steroid receptor coactivator 1 in this area. The changes in aromatase concentration presumably control seasonal variations as well as sex differences in brain estrogen production. Aromatase activity in hypothalamic homogenates is also rapidly (within minutes) down-regulated by exposure to conditions that enhance protein phosphorylation such as the presence of high concentrations of calcium, magnesium and ATP. Similarly, pharmacological manipulations such as treatment with thapsigargin or stimulation of various neurotransmitter receptors (alpha-amino-3-hydroxy-methyl-4-isoxazole propionic acid (AMPA), kainate, and N-methyl-D-aspartate (NMDA)) leading to enhanced intracellular calcium concentrations depress within minutes the aromatase activity measured in quail preoptic explants. The effects of receptor stimulation are presumably direct: electrophysiological data confirm the presence of these receptors in the membrane of aromatase-expressing cells. Inhibitors of protein kinases interfere with these processes and Western blotting experiments on brain aromatase purified by immunoprecipitation confirm that the phosphorylations regulating aromatase activity directly affect the enzyme rather than another regulatory protein. Accordingly, several phosphorylation consensus sites are present on the deduced amino acid sequence of the recently cloned quail aromatase. Fast changes in the local availability of estrogens in the brain can thus be caused by aromatase phosphorylation so that estrogen could rapidly regulate neuronal physiology and behavior. The rapid as well as slower processes of local estrogen production in the brain thus match well with the genomic and non-genomic actions of steroids in the brain. These two processes potentially provide sufficient temporal variation in the bio-availability of estrogens to support the entire range of established effects for this steroid.
Collapse
Affiliation(s)
- Jacques Balthazart
- Research Group in Behavioral Neuroendocrinology, Center for Cellular and Molecular Neurobiology, University of Liège, 17 Place Delcour (Bat L1), Liège B-4020, Belgium.
| | | | | | | | | |
Collapse
|
25
|
Abstract
My work in physiology was designed to investigate the process of androgen action within target cells and to use this information to provide insight into the clinical disorders of androgen action. The discovery that the circulating male androgen testosterone is 5alpha-reduced to a more potent hormone, dihydrotestosterone, in target tissues and that dihydrotestosterone and testosterone work by binding to the same androgen receptor protein has provided insight into the inherited syndromes of androgen resistance that impair the formation of the male urogenital tract during embryogenesis and into the role of continued dihydrotestosterone formation in the pathogenesis of prostatic hyperplasia in dogs and men.
Collapse
Affiliation(s)
- Jean D Wilson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8857, USA.
| |
Collapse
|
26
|
Shozu M, Sebastian S, Takayama K, Hsu WT, Schultz RA, Neely K, Bryant M, Bulun SE. Estrogen excess associated with novel gain-of-function mutations affecting the aromatase gene. N Engl J Med 2003; 348:1855-65. [PMID: 12736278 DOI: 10.1056/nejmoa021559] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Gynecomastia of prepubertal onset may result from increased estrogen owing to excessive aromatase activity in extraglandular tissues. A gene in chromosome 15q21.2 encodes aromatase, the key enzyme for estrogen biosynthesis. Several physiologic tissue-specific promoters regulate the expression of aromatase, giving rise to messenger RNA (mRNA) species with an identical coding region but tissue-specific 5'-untranslated regions in placenta, gonads, brain, fat, and skin. METHODS We studied skin, fat, and blood samples from a 36-year-old man, his 7-year-old son, and an unrelated 17-year-old boy with severe gynecomastia of prepubertal onset and hypogonadotropic hypogonadism caused by elevated estrogen levels. RESULTS Aromatase activity and mRNA levels in fat and skin and whole-body aromatization of androstenedione were severely elevated. Treatment with an aromatase inhibitor decreased serum estrogen levels and normalized gonadotropin and testosterone levels. The 5'-untranslated regions of aromatase mRNA contained the same sequence (FLJ) in the father and son and another sequence (TMOD3) in the unrelated boy; neither sequence was found in control subjects. These 5'-untranslated regions normally make up the first exons of two ubiquitously expressed genes clustered in chromosome 15q21.2-3 in the following order (from telomere to centromere): FLJ, TMOD3, and aromatase. The aromatase gene is normally transcribed in the direction opposite to that of TMOD3 and FLJ. Two distinct heterozygous inversions reversed the direction of the TMOD3 or FLJ promoter in the patients. CONCLUSIONS Heterozygous inversions in chromosome 15q21.2-3, which caused the coding region of the aromatase gene to lie adjacent to constitutively active cryptic promoters that normally transcribe other genes, resulted in severe estrogen excess owing to the overexpression of aromatase in many tissues.
Collapse
Affiliation(s)
- Makio Shozu
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Balthazart J, Baillien M, Charlier TD, Ball GF. Calcium-dependent phosphorylation processes control brain aromatase in quail. Eur J Neurosci 2003; 17:1591-606. [PMID: 12752377 DOI: 10.1046/j.1460-9568.2003.02598.x] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Increased gene transcription activated by the binding of sex steroids to their cognate receptors is one important way in which oestrogen synthase (aromatase) activity is regulated in the brain. This control mechanism is relatively slow (hours to days) but recent data indicate that aromatase activity in quail preoptic-hypothalamic homogenates is also rapidly (within minutes) affected by exposure to conditions that enhance Ca2+-dependent protein phosphorylation. We demonstrate here that Ca2+-dependent phosphorylations controlled by the activity of multiple protein kinases including PKC, and possibly also PKA and CAMK, can rapidly down-regulate aromatase activity in brain homogenates. These phosphorylations directly affect the aromatase molecule itself. Western blotting experiments on aromatase purified by immunoprecipitation reveal the presence on the enzyme of phosphorylated serine, threonine and tyrosine residues in concentrations that are increased by phosphorylating conditions. Cloning and sequencing of the quail aromatase identified a 1541-bp open reading frame that encodes a predicted 490-amino-acid protein containing all the functional domains that have been previously described in the mammalian and avian aromatase. Fifteen predicted consensus phosphorylation sites were identified in this sequence, but only two of these (threonine 455 and 486) match the consensus sequences corresponding to the protein kinases that were shown to affect aromatase activity during the pharmacological experiments (i.e. PKC and PKA). This suggests that the phosphorylation of one or both of these residues represents the mechanism underlying, at least in part, the rapid changes in aromatase activity.
Collapse
Affiliation(s)
- J Balthazart
- Center for Cellular and Molecular Neurobiology, Research Group in Behavioural Neuroendocrinology, University of Liège, 17 place Delcour (Bat. L1), B-4020 Liège, Belgium.
| | | | | | | |
Collapse
|
28
|
Etches RJ. From chicken coops to genome maps: generating phenotype from the molecular blueprint. Poult Sci 2001; 80:1657-61. [PMID: 11771877 DOI: 10.1093/ps/80.12.1657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The tools of molecular and cellular biology can be used to precisely describe traits in terms of a sequence of nucleic acids when their molecular and cellular bases are well understood. The entire genome of elite production birds, however, cannot be written as a series of A's, T's, C's, and G's because the interaction between alleles at the same and different loci is too large and there is likely to be many genotypes that encode the same production trait phenotype. A first draft of the genetic map of the chicken is anticipated within the next few years, but a complete molecular description of the genome of birds with elite production characteristics is not anticipated in the near future. Quantitative genetics will remain the cornerstone of breeding programs for production traits. Novel sequences encoding traits such as enhanced nutritional capability (e.g., expression of phytase) and resistance to specific diseases could be introduced into lines of chickens using the tools of molecular and cellular biology. Cloning could be used by the poultry industry to disperse highly desirable genotypes without the need for grandparent and parent flocks for multiplication.
Collapse
Affiliation(s)
- R J Etches
- Origen Therapeutics, Burlingame, California 94010, USA.
| |
Collapse
|
29
|
Gabriel WN, Blumberg B, Sutton S, Place AR, Lance VA. Alligator aromatase cDNA sequence and its expression in embryos at male and female incubation temperatures. THE JOURNAL OF EXPERIMENTAL ZOOLOGY 2001; 290:439-48. [PMID: 11555851 DOI: 10.1002/jez.1087] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In all species of crocodilians, sex is determined not by genetic mechanisms, but by the temperature at which the egg is incubated. In the American alligator (Alligator mississippiensis) the thermosensitive period (TSP) for sex determination is a 7- to 10-day window within stages 21-24 of development, around the middle third of the incubation period. Treating embryos with estrogen during the TSP produces female offspring, even at male incubation temperatures. Conversely, blocking embryonic estrogen synthesis at female-inducing temperature prevents development of the female phenotype. Therefore, it has been suggested that estrogen plays a role in determination of sex in the alligator. Estrogen is produced from an androgen substrate by cytochrome P450 aromatase (CYP19). If estrogen plays a critical role in sex determination, there should be differences in aromatase expression between embryos at male- and female-producing temperatures during the TSP. Therefore, to address this question, we cloned and characterized the alligator CYP19 cDNA. Based on the sequence information, a quantitative kinetic reverse transcriptase-polymerase chain reaction (TaqMan) assay was designed to measure expression of the alligator aromatase gene in RNA extracted from the gonadal and brain regions of alligator embryos incubated at male- or female-producing temperatures from prior to the TSP through hatching. Aromatase expression was detected in the brain region from the earliest stage tested (stage 20) through hatching. The hypothalamus had significantly higher expression than the forebrain or hindbrain in both male and female embryos. Expression was not significantly different in the gonadal region between embryos at male and female temperatures until after the TSP, when there was a dramatic increase in expression at female temperature. These data indicate that aromatase expression and, thus, estrogen production, are not the initial trigger for sex determination but play an essential role in ovarian differentiation in the alligator. J. Exp. Zool. 290:439-448, 2001.
Collapse
Affiliation(s)
- W N Gabriel
- Center for Reproduction of Endangered Species, San Diego, California 92101, USA
| | | | | | | | | |
Collapse
|
30
|
Lanzino M, Catalano S, Genissel C, Ando S, Carreau S, Hamra K, McPhaul MJ. Aromatase messenger RNA is derived from the proximal promoter of the aromatase gene in Leydig, Sertoli, and germ cells of the rat testis. Biol Reprod 2001; 64:1439-43. [PMID: 11319149 DOI: 10.1095/biolreprod64.5.1439] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
It has long been recognized that individual cell types within the testes possess the capacity to synthesize estrogen. A number of studies on different species have demonstrated that the levels of aromatase expression and the patterns of regulation are distinct between the different cell types of the testes. Whereas a variety of promoters have been shown to contribute to the patterns of aromatase expression in different cell lineages, studies using ovarian RNA, testis RNA, and Leydig cell tumor lines have demonstrated that the same promoter (promoter II) was used in each. Recent experiments using potent aromatase inhibitors or analysis of animals in which the genes encoding the estrogen receptor-alpha (ER-alpha) or the aromatase, P450, are defective, have confirmed the importance of local estrogen formation in normal testicular function. In order to permit experiments to identify the elements controlling aromatase expression in the individual cell compartments of the testes, we prepared RNA from purified preparations of Leydig, Sertoli, and germ cells. Using specific oligonucleotide primers, the sites of initiation of the aromatase mRNA were determined using rapid amplification of cDNA ends (RACE) and nucleotide sequence analysis of the resulting cDNA fragments. Our results indicate that aromatase mRNA is derived from the proximal promoter (PII) of the aromatase gene in each of the major cell types of the rat testes.
Collapse
Affiliation(s)
- M Lanzino
- Department of Cell Biology, Faculty of Pharmacy, University of Calabria, 87030 Rende (CS), Italy
| | | | | | | | | | | | | |
Collapse
|
31
|
Melo AC, Ramsdell JS. Sexual dimorphism of brain aromatase activity in medaka: induction of a female phenotype by estradiol. ENVIRONMENTAL HEALTH PERSPECTIVES 2001; 109:257-64. [PMID: 11333187 PMCID: PMC1240244 DOI: 10.1289/ehp.01109257] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In this study we identified sex-dependent dimorphism of brain aromatase in the teleost medaka and examined its regulation by sex steriods. We first investigated differential distribution of brain aromatase activity in sexually mature male and female medaka in serial coronal sections of the brain and identified the hypothalamic nuclei contained in each section using the brain atlas of medaka. In the brain of male medaka, high levels of activity are localized in sections containing the preoptic (POA) and suprachiasmatic nuclei (SC) (63-75 fmol/hr) and low levels in the nuclei periventricular dorsalis (HD), ventralis (HV), and caudalis (Hc), nuclei diffusus of lobulus inferiores (NDIL), and nuclei tuberi anteriores (TA) and posteriores (TP) (< 25 fmol/hr). In the brain of female medaka high aromatase activity is localized in sections containing the HD, HV, Hc, NDIL, TA, and TP (85-80 fmol/hr) and highly variable levels in the POA and SC (23-70 fmol/hr). The concentration and time dependency of the exposure of male medaka to estradiol on the total brain aromatase activity and morphologic sex characteristics were determined next. Estradiol increased the activity of brain aromatase in a concentration-dependent manner at 2.5 and 25 microg/L, but the increase was lower at higher concentrations of the hormone. The effect was time dependent, gradually increasing up to the fifth day of exposure, after which it reached a plateau. Estradiol induction of brain aromatase analyzed using Lineweaver-Burke plots of saturation assays revealed a non-first-order reaction. The results indicate that a positive feedback mechanism regulates brain aromatase and imply that the sexual dimorphic distribution of aromatase may be highly sensitive to physiologic cues and environmental perturbations in fish.
Collapse
Affiliation(s)
- A C Melo
- Marine Biotoxins Program, Center for Coastal Environmental Health and Biomolecular Research, National Atmospheric and Oceanic Administration-National Ocean Service, Charleston, SC 29412, USA
| | | |
Collapse
|
32
|
Miyashita K, Shimizu N, Osanai S, Miyata S. Sequence analysis and expression of the P450 aromatase and estrogen receptor genes in the Xenopus ovary. J Steroid Biochem Mol Biol 2000; 75:101-7. [PMID: 11226826 DOI: 10.1016/s0960-0760(00)00164-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recent studies point to a key role for the estrogen synthesizing enzyme P450 aromatase (P450 arom) in ovary determination in fish, birds and reptiles. It is unclear whether estrogen synthesis is important in sex determination of Xenopus gonad. To determine whether the aromatase gene is transcribed in the gonads of Xenopus tadpoles during the sex determination, we cloned a P450 arom cDNA and examined the level of P450 arom and estrogen receptor (ER) gene expression in association with estrogen activity. cDNA clones for P450 arom were isolated from a Xenopus ovarian cDNA library. There was an open reading frame (ORF) of 1500 bp from the ATG start to TAA stop codons encoding 500 predicted amino acids. cDNAs for P450 arom have previously been cloned from various vertebrates. The homology between the Xenopus P450 aromatase and the human P450 arom was higher. The expression of the P450 arom gene was mainly limited to reproductive organs. To determine the beginning of estrogen activity in gonads of embryos, expression of the aromatase and ER gene was also examined by RQ-RT-PCR. Both Xenopus aromatase and ER mRNA was detected at stage 51 in gonads. These observations are consistent with estrogens having a key role in ovarian development in various other vertebrates.
Collapse
Affiliation(s)
- K Miyashita
- Institute for Advanced Medical Research, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | |
Collapse
|
33
|
Ramachandran B, Schlinger BA, Arnold AP, Campagnoni AT. Zebra finch aromatase gene expression is regulated in the brain through an alternate promoter. Gene 1999; 240:209-16. [PMID: 10564828 DOI: 10.1016/s0378-1119(99)00399-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The learned singing behavior in songbirds is sex-steroid-dependent and sexually dimorphic. Estrogen plays a major role in masculinizing the song system in these songbirds. The songbird brain synthesizes large amounts of estrogen, which, in the case of zebra finches, have been found to enter the systemic circulation. Aromatase cytochrome P450 is the key enzyme catalyzing the conversion of androgens to estrogens. We have cloned a novel alternatively spliced form of aromatase cDNA expressed predominantly in the zebra finch brain. We have also isolated and characterized the gene coding for zebra finch aromatase which spans 20kb in length. The alternate forms of aromatase mRNA (ARO) differ in their 5'-untranslated regions encoded by either exon 1a or 1b. The putative promoter sequences controlling the regulation of the alternate forms of ARO in zebra finches contain consensus binding sites for various transcription factors. While both the promoters have binding sites for SRY-like transcription factor, a binding site for SF-1 is present only in the promoter 1b active in the ovary. Intriguingly, a 55bp segment within the promoter 1a sequence appears to be highly conserved among zebra finch, mouse and human aromatase promoters active in the brain.
Collapse
Affiliation(s)
- B Ramachandran
- Departments of Psychiatry, Laboratory of Neuroendocrinology of the Brain Research Institute, Mental Retardation Research Center, University of California, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
34
|
Brosius J. RNAs from all categories generate retrosequences that may be exapted as novel genes or regulatory elements. Gene 1999; 238:115-34. [PMID: 10570990 DOI: 10.1016/s0378-1119(99)00227-9] [Citation(s) in RCA: 275] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
While the significance of middle repetitive elements had been neglected for a long time, there are again tendencies to ascribe most members of a given middle repetitive sequence family a functional role--as if the discussion of SINE (short interspersed repetitive elements) function only can occupy extreme positions. In this article, I argue that differences between the various classes of retrosequences concern mainly their copy numbers. Consequently, the function of SINEs should be viewed as pragmatic such as, for example, mRNA-derived retrosequences, without underestimating the impact of retroposition for generation of novel protein coding genes or parts thereof (exon shuffling by retroposition) and in particular of SINEs (and retroelements) in modulating genes and their expression. Rapid genomic change by accumulating retrosequences may even facilitate speciation [McDonald, J.F., 1995. Transposable elements: possible catalysts of organismic evolution. Trends Ecol. Evol. 10, 123-126.] In addition to providing mobile regulatory elements, small RNA-derived retrosequences including SINEs can, in analogy to mRNA-derived retrosequences, also give rise to novel small RNA genes. Perhaps not representative for all SINE/master gene relationships, we gained significant knowledge by studying the small neuronal non-messenger RNAs, namely BC1 RNA in rodents and BC200 RNA in primates. BC1 is the first identified master gene generating a subclass of ID repetitive elements, and BC200 is the only known Alu element (monomeric) that was exapted as a novel small RNA encoding gene.
Collapse
Affiliation(s)
- J Brosius
- Institute of Experimental Pathology/Molecular Neurobiology, ZMBE, University of Münster, Germany.
| |
Collapse
|
35
|
Goldstein JL, Brown MS. Presentation of the Kober Medal for 1999 to Jean D. Wilson physician-scientist exemplar. PROCEEDINGS OF THE ASSOCIATION OF AMERICAN PHYSICIANS 1999; 111:469-79. [PMID: 10519168 DOI: 10.1111/paa.1999.111.5.469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- J L Goldstein
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas 75235-9046, USA.
| | | |
Collapse
|
36
|
Kimball RT, Ligon JD. Evolution of Avian Plumage Dichromatism from a Proximate Perspective. Am Nat 1999. [DOI: 10.1086/303228] [Citation(s) in RCA: 124] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
37
|
Young M, McPhaul MJ. A steroidogenic factor-1-binding site and cyclic adenosine 3',5'-monophosphate response element-like elements are required for the activity of the rat aromatase promoter in rat Leydig tumor cell lines. Endocrinology 1998; 139:5082-93. [PMID: 9832447 DOI: 10.1210/endo.139.12.6377] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although transcription initiation within CYP19 (cytochrome P450 aromatase) occurs immediately 5' to the initiator methionine (proximal promoter) in two rat Leydig tumor cell lines (R2C and H540) that express high aromatase activity and in rat ovary, the patterns of aromatase expression in the two cell types are distinctive. To define mechanisms controlling different patterns of expression of the rat aromatase proximal promoter, we performed transient transfection and gel mobility shift assays. Transfection experiments using different sized promoter fragments fused to a reporter gene were used to identify regions that are functionally important for transcriptional regulation in steroidogenic cell lines [R2C, H540, and Y1 (mouse adrenocortical cells that express low aromatase activity)]. These experiments indicate that the cAMP response element (CRE) at -231 and the steroidogenic factor-1 (SF1) motif are both required for expression of the reporter gene in each steroidogenic cell line and that the CRE at -169 is similarly required in R2C cells. Gel mobility shift assays confirm binding of nuclear proteins from the steroidogenic cell lines to the SF1 motif and to CRE (-231). Leydig tumor cells also contain nuclear proteins that bind to the CRE (-169), but nuclear extracts from R2C cells produce a uniquely shifted band compared with H540 cells. These results suggest that differences in proteins that bind to distinct elements within the rat aromatase promoter may be responsible for different patterns and levels of aromatase expression in these steroidogenic cell lines.
Collapse
Affiliation(s)
- M Young
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas 75235-8857, USA
| | | |
Collapse
|
38
|
Gupta RP, Abou-Donia MB. Cytochrome P450 enzymes in chickens: characteristics and induction by xenobiotics. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART C, PHARMACOLOGY, TOXICOLOGY & ENDOCRINOLOGY 1998; 121:73-83. [PMID: 9972452 DOI: 10.1016/s0742-8413(98)10031-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- R P Gupta
- Department of Pharmacology, Duke University Medical Center, Durham, NC 27708, USA
| | | |
Collapse
|
39
|
|
40
|
Tchoudakova A, Callard GV. Identification of multiple CYP19 genes encoding different cytochrome P450 aromatase isozymes in brain and ovary. Endocrinology 1998; 139:2179-89. [PMID: 9529008 DOI: 10.1210/endo.139.4.5899] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Evidence to date indicates that the gene encoding cytochrome P450 aromatase (P450arom) in humans is a single member of the CYPl9 family, but multiple CYPl9 loci and isoforms have been identified in pigs. Here we report the cloning and characterization of a second member of the CYP19 family in goldfish. A search for P450arom variants was prompted by studies showing that a full-length P450arom complementary DNA (cDNA) isolated from a goldfish brain cDNA library hybridizes with a high abundance 3 kb transcript in brain RNA but fails to detect a message in ovarian RNA. A stepwise PCR cloning strategy led to isolation of a 1.9-kb cDNA, which encodes a protein of 518 amino acids and has a predicted mol wt of 58.7K. The ovary-derived P450arom (-A) shares 68-72% sequence identity with ovarian aromatases of other fish species, but only 62% identity with the homologous brain-derived P450arom (-B). Amino acid differences are distributed throughout the two goldfish P450arom forms, but presumptive functional domains are highly conserved. Both P450aromA and -B are able to aromatize [3H]androgen to [3H]estrogen when expressed in nonsteroidogenic COS cells. Southern analysis and PCR-restriction analysis of genomic DNA using discriminating probes and primers indicates that a single locus encodes the brain-derived P450aromB (CYPl9B), whereas one or two different loci encode the ovarian form (CYPl9A). Northern blot analysis revealed two P450aromA messenger RNAs (1.9 >> 3.0 kb) in ovary. Simultaneous PCR amplification with A- and B-specific primer pairs confirms that P450aromA is the only form expressed in ovaries, but shows overlapping expression of the two genes in neural tissues. Whereas P450aromB messenger RNA predominates in brain (B/A, approximately 14:1), the ratios are reversed in retina (B/A, approximately 1:25). Further studies are required to resolve the evolutionary and functional implications of multiple CYPl9 genes and P450arom isozymes in goldfish, their differential expression in brain and ovary, and whether observations can be generalized to other vertebrates.
Collapse
Affiliation(s)
- A Tchoudakova
- Department of Biology, Boston University, Massachusetts 02215, USA
| | | |
Collapse
|
41
|
Gelinas D, Pitoc GA, Callard GV. Isolation of a goldfish brain cytochrome P450 aromatase cDNA: mRNA expression during the seasonal cycle and after steroid treatment. Mol Cell Endocrinol 1998; 138:81-93. [PMID: 9685217 DOI: 10.1016/s0303-7207(98)00015-x] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To investigate the molecular basis and physiological regulation of exceptionally high levels of aromatase (P450arom) activity in the brain of teleost fish, a 2927 bp P450arom cDNA encoding a 510 amino acid protein was isolated from a goldfish brain cDNA library. The brain-derived cDNA had 53% and 61-62% sequence identity when compared with human placental and fish ovarian P450arom forms, respectively, and higher homologies in conserved functional domains. Goldfish brain poly(A) RNA was translatable in vitro to a 56 kDa P450arom immunoprecipitation product. Northern blot analysis using the brain cDNA revealed a major 3.0 kb transcript of high abundance in brain (FB, forebrain > M/HB, mid/hindbrain), but no signal in ovary, testis or liver. P450arom mRNA varied seasonally in brain, with a peak at the onset of gonadal regrowth (February) that preceded the annual rise in enzyme levels and was 4-fold (FB) or 50-fold (M/HB) higher than during reproductive inactivity (July-December). Known markers of neurogenesis and estrogen action in brain (28S rRNA, beta-actin and beta-tubulin transcripts) each had unique seasonal patterns which differed from P450arom mRNA. In vivo steroid treatment showed that estrogen and aromatizable androgen increase FB and M/HB levels of P450arom mRNA 8- and 4-fold, respectively. P450arom mRNA in pituitary and retina had a different regulation. Southern analysis provided no evidence for multiple genes encoding the brain derived cDNA or for brain-specific gene amplification. Results imply that high accumulated levels of P450arom mRNA are the major determinant of high measured enzyme activity in goldfish brain, and that physiological regulation of mRNA expression in the natural environment is mediated by aromatization of androgen to estrogen.
Collapse
Affiliation(s)
- D Gelinas
- Department of Biology, Boston University, MA 02215, USA
| | | | | |
Collapse
|
42
|
Abstract
Sexual dimorphism in humans has been the subject of wonder for centuries. In 355 BC, Aristotle postulated that sexual dimorphism arose from differences in the heat of semen at the time of copulation. In his scheme, hot semen generated males, whereas cold semen made females (Jacquart, D., and C. Thomasset. Sexuality and Medicine in the Middle Ages, 1988). In medieval times, there was great controversy about the existence of a female pope, who may have in fact had an intersex phenotype (New, M. I., and E. S. Kitzinger. J. Clin. Endocrinol. Metab. 76: 3-13, 1993.). Recent years have seen a resurgence of interest in mechanisms controlling sexual differentiation in mammals. Sex differentiation relies on establishment of chromosomal sex at fertilization, followed by the differentiation of gonads, and ultimately the establishment of phenotypic sex in its final form at puberty. Each event in sex determination depends on the preceding event, and normally, chromosomal, gonadal, and somatic sex all agree. There are, however, instances where chromosomal, gonadal, or somatic sex do not agree, and sexual differentiation is ambiguous, with male and female characteristics combined in a single individual. In humans, well-characterized patients are 46, XY women who have the syndrome of pure gonadal dysgenesis, and a subset of true hermaphrodites are phenotypic men with a 46, XX karyotype. Analysis of such individuals has permitted identification of some of the molecules involved in sex determination, including SRY (sex-determining region Y gene), which is a Y chromosomal gene fulfilling the genetic and conceptual requirements of a testis-determining factor. The purpose of this review is to summarize the molecular basis for syndromes of sexual ambiguity seen in human patients and to identify areas where further research is needed. Understanding how sex-specific gene activity is orchestrated may provide insight into the molecular basis of other cell fate decisions during development which, in turn, may lead to an understanding of aberrant cell fate decisions made in patients with birth defects and during neoplastic change.
Collapse
Affiliation(s)
- C M Haqq
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, USA
| | | |
Collapse
|
43
|
Andrews JE, Smith CA, Sinclair AH. Sites of estrogen receptor and aromatase expression in the chicken embryo. Gen Comp Endocrinol 1997; 108:182-90. [PMID: 9356214 DOI: 10.1006/gcen.1997.6978] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Estrogens have been implicated in sexual differentiation of both the gonads and the genitalia of birds. In chicken embryos, the gonads are steroidogenically active from an early age, and the aromatase gene, (cAROM), necessary for estrogen synthesis, is expressed only in females at the time of gonadal sex differentiation. However, no studies have directly demonstrated the distribution of estrogen receptor (cER) transcripts or proteins in the embryonic avian reproductive system. Whole-mount in situ hybridization and immunohistochemistry were used here to identify sites of estrogen receptor expression in the embryonic chicken urogenital system. Estrogen receptor mRNA was observed in both male and female gonads prior to morphological differentiation, at Stage 26 (4.5 days of incubation), and continued until after sexual dimorphism at Stage 32 (7.5 days). Transcripts of cER were also detected in the Müllerian ducts and developing external genitalia of both sexes. Estrogen receptor protein was analysed in the embryonic gonads by immunohistochemistry and found to be most abundant in the cortex of the left ovary, although it was also present in the medulla of both female gonads. No significant cER protein expression was detected in the male gonad by immunohistochemistry. In contrast, the aromatase gene was expressed in the gonads of female embryos from the onset of sexual dimorphism but was not detectable in male gonads at any stage examined. These findings suggest that estrogen involvement in both gonadogenesis and genital development in chickens is mediated by the estrogen receptor.
Collapse
Affiliation(s)
- J E Andrews
- Centre for Hormone Research, The University of Melbourne, Parkville, Victoria 3052, Australia
| | | | | |
Collapse
|
44
|
Abstract
The rat H540 Leydig cell tumor has been shown to express cholesterol side-chain cleavage and 17alpha-hydroxylase cytochrome P450s, 3beta-hydroxysteroid dehydrogenase/delta 5-delta 4 isomerase, and steroid 5alpha-reductase, making it a useful model in which to study steroidogenesis. In the current studies, we report that cultured H540 cells express high levels of aromatase cytochrome P450 (P450arom), which converts androgens to estrogens. Levels of aromatase activity varied from 9.4 to 51.7 pmol/h/mg protein and inhibition of 5alpha-reductase with finasteride did not significantly effect aromatase measurements, indicating that 5alpha-reductase is not competing for the substrate used in the aromatase assays. Aromatase activity was decreased 95% by preincubating the cells with 4-hydroxyandrostenedione, an aromatase inhibitor. Characterization of the aromatase mRNA expressed in the H540 cell line demonstrates that, like R2C cells and rat ovarian tissue, three distinct P450arom mRNA species are detected by Northern analysis, and that these transcripts are derived from the same site of transcription initiation. Despite these similarities, the regulation of aromatase activity by 8-bromo-cAMP in H540 cells differs from both R2C cells and rat ovarian tissue. As the H540 and R2C cell lines appear to have distinct origins, H540 is the second rat Leydig tumor cell line characterized that constitutively expresses high levels of aromatase.
Collapse
Affiliation(s)
- M Young
- Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, 75235-8857, USA
| | | | | |
Collapse
|
45
|
Choi I, Troyer DL, Cornwell DL, Kirby-Dobbels KR, Collante WR, Simmen FA. Closely related genes encode developmental and tissue isoforms of porcine cytochrome P450 aromatase. DNA Cell Biol 1997; 16:769-77. [PMID: 9212170 DOI: 10.1089/dna.1997.16.769] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
We examined the chromosomal basis for the synthesis of tissue (ovary, endometrium/placenta, and peri-implantation blastocyst) isoforms of cytochrome P450 aromatase in the pig. DNA fragments derived from three distinct porcine aromatase chromosomal genes were cloned and characterized. The porcine type III aromatase gene encoding the blastocyst aromatase isoform was found to consist of nine coding exons and two mutually exclusive, 5' untranslated exons (designated E1A and E1B), collectively spanning 30 kb or more. The porcine type II aromatase gene, encoding the endometrial/placental aromatase isoform, was identified by cloning of a genomic DNA fragment spanning the corresponding exons 7, 8, and 9. The DNA inserts of two other phage clones encompassed exons 2, 3, and 4 of a third chromosomal gene (type I) encoding the ovarian aromatase isoform. All intron-exon junctions in these genomic fragments were found to be identical in relative positions to those of the single-copy human aromatase gene. Comparisons of cDNA and genomic sequences indicated that nucleotide sequence variation was not uniform across the corresponding exons of these genes and that the corresponding intronic sequences were conserved. The type II and type III aromatase genes were localized to the same regional location (q16-17) on swine chromosome 1, which is homologous to the human chromosome 15 region (q21.1) in which the human aromatase gene resides. Results demonstrate that the three aromatase genes characterized in the present study appear to be similar in their overall structural organization and most likely are clustered, which could have resulted from at least two independent gene duplication events. The presence of multiple aromatase genes constitutes a newly described mechanism by which aromatase enzyme biosynthesis and functional activity can be regulated in a tissue and temporal fashion and serves to highlight further the complexity of aromatase gene expression in mammals. Moreover, the presence of a unique aromatase gene that is highly expressed in pig blastocysts may constitute a paradigm for other mammals (e.g., equids, rabbit, hamster) whose peri-implantation blastocysts are estrogenic.
Collapse
Affiliation(s)
- I Choi
- Department of Dairy and Poultry Sciences and the Interdisciplinary Concentration in Animal Molecular and Cell Biology, Institute of Food and Agricultural Sciences, University of Florida, Gainesville 32611-0920, USA
| | | | | | | | | | | |
Collapse
|
46
|
Smith CA, Andrews JE, Sinclair AH. Gonadal sex differentiation in chicken embryos: expression of estrogen receptor and aromatase genes. J Steroid Biochem Mol Biol 1997; 60:295-302. [PMID: 9219920 DOI: 10.1016/s0960-0760(96)00196-3] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Estrogen is implicated in sexual differentiation of the avian gonad. Expression of the estrogen receptor and aromatase genes was therefore examined at the time of gonadal sex differentiation in chicken embryos, using reverse transcription and the polymerase chain reaction (RT-PCR). Estrogen receptor (cER) transcripts were detected in the gonads of both presumptive sexes at embryonic days 4.5, 5.5 and 6.5, and in female but not male urogenital tissues at day 3.5. Aromatase (cAROM) transcripts were detected in female but not male gonads from day 6.5 of embryogenesis, and in adult gonads of both sexes. Both female and male embryos thus express cER mRNA before morphological differentiation of the gonads, which begins on day 5, whereas cAROM expression begins at or shortly after the onset of differentiation and is female-specific. Examination of other tissues showed that, in 5.5-day-old embryos, cER expression was limited to the gonads; no transcripts were detected in the mesonephric kidney, liver, brain, hindlimb or heart of either sex. In 9.5-day-old female embryos, cER and cAROM transcripts were present in both the left (ovarian) and the right (regressing) gonads. Altogether, these observations imply that the gonads of both sexes develop the capacity to respond to estrogens early in embryogenesis, before morphological differentiation, whereas the capacity to synthesize estrogens is female-specific and occurs later, at the time of differentiation. These observations are consistent with estrogens having a key role in ovarian development.
Collapse
Affiliation(s)
- C A Smith
- Department of Paediatrics and Centre for Hormone Research, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia
| | | | | |
Collapse
|
47
|
KUDO T, YAMAMOTO H, SATO S, SUTOU S. Comparison of 5' Upstream Regions of Chicken and Quail Aromatase Genes. J Reprod Dev 1996. [DOI: 10.1262/jrd.42.101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Toshiyuki KUDO
- Central Research Institute, Itoham Foods Inc., 1-2, Kubogaoka, Moriya, Kitasouma, Ibaraki 302-01, Japan
| | - Hiroaki YAMAMOTO
- Biological Institute, Faculty of Science, Tohoku University, Sendai 980, Japan
| | - Seiji SATO
- Central Research Institute, Itoham Foods Inc., 1-2, Kubogaoka, Moriya, Kitasouma, Ibaraki 302-01, Japan
| | - Shizuyo SUTOU
- Central Research Institute, Itoham Foods Inc., 1-2, Kubogaoka, Moriya, Kitasouma, Ibaraki 302-01, Japan
| |
Collapse
|
48
|
Abstract
Reverse transcription has been an important mediator of genomic change. This influence dates back more than three billion years, when the RNA genome was converted into the DNA genome. While the current cellular role(s) of reverse transcriptase are not yet completely understood, it has become clear over the last few years that this enzyme is still responsible for generating significant genomic change and that its activities are one of the driving forces of evolution. Reverse transcriptase generates, for example, extra gene copies (retrogenes), using as a template mature messenger RNAs. Such retrogenes do not always end up as nonfunctional pseudogenes but form, after reinsertion into the genome, new unions with resident promoter elements that may alter the gene's temporal and/or spatial expression levels. More frequently, reverse transcriptase produces copies of nonmessenger RNAs, such as small nuclear or cytoplasmic RNAs. Extremely high copy numbers can be generated by this process. The resulting reinserted DNA copies are therefore referred to as short interspersed repetitive elements (SINEs). SINEs have long been considered selfish DNA, littering the genome via exponential propagation but not contributing to the host's fitness. Many SINEs, however, can give rise to novel genes encoding small RNAs, and are the migrant carriers of numerous control elements and sequence motifs that can equip resident genes with novel regulatory elements [Brosius J. and Gould S.J., Proc Natl Acad Sci USA 89, 10706-10710, 1992]. Retrosequences, such as SINEs and portions of retroelements (e.g., long terminal repeats, LTRs), are capable of donating sequence motifs for nucleosome positioning, DNA methylation, transcriptional enhancers and silencers, poly(A) addition sequences, determinants of RNA stability or transport, splice sites, and even amino acid codons for incorporation into open reading frames as novel protein domains. Retroposition can therefore be considered as a major pacemaker for evolution (including speciation). Retroposons, with their unique properties and actions, form the molecular basis of important evolutionary concepts, such as exaptation [Gould S.J. and Vrba E., Paleobiology 8, 4-15, 1982] and punctuated equilibrium [Elredge N. and Gould S.J. in Schopf T.J.M. (ed). Models in Paleobiology. Freeman, Cooper, San Francisco, 1972, pp. 82-115].
Collapse
Affiliation(s)
- J Brosius
- Institute for Experimental Pathology, ZMBE University of Münster, Germany.
| | | |
Collapse
|
49
|
Lephart ED, Herbst MA, McPhaul MJ. Characterization of aromatase cytochrome P-450 mRNA in rat perinatal brain, ovary and a Leydig tumor cell line: evidence for the existence of brain specific aromatase transcripts. Endocrine 1995; 3:25-31. [PMID: 21153233 DOI: 10.1007/bf02917445] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/1994] [Accepted: 09/07/1994] [Indexed: 10/21/2022]
Abstract
The conversion of androgens to estrogens is catalyzed by the aromatase cytochrome P-450 (P-450(AROM)) in a veriety of tissues and cell types in vertebrates. The manner in which aromatase activity is regulated appears to be quite different, even between different tissues of a single species. In the current study, we have determined the sequence of the 5' end of the aromatase mRNA in a rat Leydig tumor cell line, R2C, and in three rat tissue [adult ovary, perinatal amygdala (AmY), and medial basal hypothalamus-preoptic area (MBH-POA)], each of which shows different patterns of aromatase expression. S(1) nuclease protection and primer-extension analyses establish that the site of transcription initiation of the aromatase mRNA present in rat ovarh and the rat Leydig tumor cell line R2C is located approximately 97 nucleotides upstream from the initiator methionine. By contrast, although aromatase mRNA was detected in S(1) nuclease protection experiments using a probe derived from the aromatase open-reading frame, transcripts initiating at this site were absent from RNA samples prepared from perinatal rat AMY and MBH-POA tissue. S(1) mapping and sequencing of the 5' end of AMY and MBH-POA aromatase cDNAs indicate that the aromatase mRNA transcripts present in these rat neural tissues from perinatal animals contain a distinctive 5' terminus and are derived from a different promoter.
Collapse
Affiliation(s)
- E D Lephart
- Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, 75235-8857, Dallas, Texas, USA
| | | | | |
Collapse
|
50
|
Ding D, Lipshitz HD. Spatially regulated expression of retrovirus-like transposons during Drosophila melanogaster embryogenesis. Genet Res (Camb) 1994; 64:167-81. [PMID: 7698641 DOI: 10.1017/s0016672300032833] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Over twenty distinct families of long terminal direct repeat (LTR)-containing retrotransposons have been identified in Drosophila melanogaster. While there have been extensive analyses of retrotransposon transcription in cultured cells, there have been few studies of the spatial expression of retrotransposons during normal development. Here we report a detailed analysis of the spatial expression patterns of fifteen families of retrotransposons during Drosophila melanogaster embryogenesis (17.6, 297, 412, 1731, 3S18, blood, copia, gypsy, HMS Beagle, Kermit/flea, mdg1, mdg3, opus, roo/B104 and springer). In each case, analyses were carried out in from two to four wild-type strains. Since the chromosomal insertion sites of any particular family of retrotransposons vary widely among wild-type strains, a spatial expression pattern that is conserved among strains is likely to have been generated through interaction of host transcription factors with cis-regulatory elements resident in the retrotransposons themselves. All fifteen families of retrotransposons showed conserved patterns of spatially and temporally regulated expression during embryogenesis. These results suggest that all families of retrotransposons carry cis-acting elements that control their spatial and temporal expression patterns. Thus, transposition of a retrotransposon into or near a particular host gene-possibly followed by an excision event leaving behind the retrotransposon's cis-regulatory sequences-might impose novel developmental control on such a host gene. Such a mechanism would serve to confer evolutionarily significant alterations in the spatio-temporal control of gene expression.
Collapse
Affiliation(s)
- D Ding
- Division of Biology 156-29, California Institute of Technology, Pasadena 91125
| | | |
Collapse
|