1
|
Wallis M, Xu Q, Krawczyk M, Skowronska-Krawczyk D. Evolution of the enhancer-rich regulatory region of the gene for the cell-type specific transcription factor POU1F1. Heliyon 2024; 10:e28640. [PMID: 38590853 PMCID: PMC10999999 DOI: 10.1016/j.heliyon.2024.e28640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Precise spatio-temporal expression of genes in organogenesis is regulated by the coordinated interplay of DNA elements such as promoter and enhancers present in the regulatory region of a given locus. POU1F1 transcription factor plays a crucial role in the development of somatotrophs, lactotrophs and thyrotrophs in the anterior pituitary gland, and in maintaining high expression of growth hormone, prolactin and TSH. In mouse, expression of POU1F1 is controlled by a region fenced by two CTCF sites, containing 5 upstream enhancer elements, designated E-A (5' to 3'). Elements C, B and A correspond to elements shown previously to play a role in pituitary development and hormonal expression; functional roles for elements E and D have not been reported. We performed comparative sequence analysis of this regulatory region and discovered that three elements, B, C and E, are present in all vertebrate groups except Agnatha. One very long (>2 kb) element (A) is unique to mammals suggesting a specific change in regulation of the gene in this group. Using DNA accessibility assay (ATAC-seq) we showed that conserved elements in anterior pituitary of four non-mammals are open, suggesting functionality as regulatory elements. We showed that, in many non-mammalian vertebrates, an additional upstream exon closely follows element E, leading to alternatively spliced transcripts. Here, element E functions as an alternative promoter, but in mammals this feature is lost, suggesting conversion of alternative promoter to enhancer. Our work shows that regulation of POU1F1 changed markedly during the course of vertebrate evolution, use of a low number of enhancer elements combined with alternative promoters in non-mammalian vertebrates being replaced by use of a unique combination of regulatory units in mammals. Most importantly, our work suggests that evolutionary conversion of alternate promoter to enhancer could be one of the evolutionary mechanisms of enhancer birth.
Collapse
Affiliation(s)
- Michael Wallis
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Qianlan Xu
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| | - Michal Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
2
|
LaPierre MP, Godbersen S, Torres Esteban M, Schad AN, Treier M, Ghoshdastider U, Stoffel M. MicroRNA-7a2 Regulates Prolactin in Developing Lactotrophs and Prolactinoma Cells. Endocrinology 2021; 162:6009069. [PMID: 33248443 PMCID: PMC7774778 DOI: 10.1210/endocr/bqaa220] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 02/06/2023]
Abstract
Prolactin production is controlled by a complex and temporally dynamic network of factors. Despite this tightly coordinated system, pathological hyperprolactinemia is a common endocrine disorder that is often not understood, thereby highlighting the need to expand our molecular understanding of lactotroph cell regulation. MicroRNA-7 (miR-7) is the most highly expressed miRNA family in the pituitary gland and the loss of the miR-7 family member, miR-7a2, is sufficient to reduce prolactin gene expression in mice. Here, we used conditional loss-of-function and gain-of-function mouse models to characterize the function of miR-7a2 in lactotroph cells. We found that pituitary miR-7a2 expression undergoes developmental and sex hormone-dependent regulation. Unexpectedly, the loss of mir-7a2 induces a premature increase in prolactin expression and lactotroph abundance during embryonic development, followed by a gradual loss of prolactin into adulthood. On the other hand, lactotroph development is delayed in mice overexpressing miR-7a2. This regulation of lactotroph function by miR-7a2 involves complementary mechanisms in multiple cell populations. In mouse pituitary and rat prolactinoma cells, miR-7a2 represses its target Raf1, which promotes prolactin gene expression. These findings shed light on the complex regulation of prolactin production and may have implications for the physiological and pathological mechanisms underlying hyperprolactinemia.
Collapse
Affiliation(s)
- Mary P LaPierre
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Svenja Godbersen
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | | | - Anaïs Nura Schad
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Mathias Treier
- Max Delbrück Zentrum für molekulare Medizin (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Germany
| | | | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
- Medical Faculty, University of Zürich, Zürich, Switzerland
- Correspondence: Markus Stoffel, Swiss Federal Institute of Technology, ETH Zürich, Institute for Molecular Health Science, HPL H36, Otto-Stern Weg 7, CH 8093 Zürich, Switzerland.
| |
Collapse
|
3
|
Zhao D, Han X, Huang L, Wang J, Zhang X, Jeon JH, Zhao Q, Dong JT. Transcription factor ZFHX3 regulates calcium influx in mammary epithelial cells in part via the TRPV6 calcium channel. Biochem Biophys Res Commun 2019; 519:366-371. [PMID: 31519324 DOI: 10.1016/j.bbrc.2019.08.148] [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] [Received: 08/21/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022]
Abstract
Zinc finger homeobox 3 (ZFHX3) is a transcription factor that regulates multiple cellular processes including cell proliferation, differentiation and neoplastic development. It is also involved in the function of steroid hormones estrogen and progesterone and the peptide hormone prolactin in mammary epithelial cells. In this study, we investigated whether and how ZFHX3 regulates intracellular calcium homeostasis in mammary epithelial cells. We found that ZFHX3 affected both store operated calcium entry and store independent calcium entry (SOCE and SICE). Simultaneously, the expression of the calcium channel TRPV6 was regulated by ZFHX3, as demonstrated by expression analysis and luciferase reporter assay. In cells with knockdown of ZFHX3, calcium entry was partially rescued by the overexpression of wild type but not the pore mutants of TRPV6. In addition, overexpression of TRPV6 promoted differentiation of the MCF10A mammary epithelial cells in three-dimensional culture, which is consistent with our previous findings that ZFHX3 is essential for mammary gland differentiation. These findings suggest that ZFHX3 plays an important role in intracellular calcium homeostasis in mammary epithelial cells, at least in part, by regulating TRPV6.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xueying Han
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lili Huang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jianpeng Wang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xi Zhang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ju-Hong Jeon
- Department of Physiology, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Qiang Zhao
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China; Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
4
|
Wang K, Hui Y, Zhang S, Wang M, Yan H, Zhu H, Qu L, Lan X, Pan C. A deletion mutation within the ATBF1 gene is strongly associated with goat litter size. Anim Biotechnol 2019; 31:174-180. [DOI: 10.1080/10495398.2018.1561459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ke Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yiqing Hui
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shaoli Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ming Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hailong Yan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, China
- Life Science Research Center, Yulin University, Yulin, China
| | - Haijing Zhu
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, China
- Life Science Research Center, Yulin University, Yulin, China
| | - Lei Qu
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, China
- Life Science Research Center, Yulin University, Yulin, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuanying Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
5
|
Wei Z, Wang K, Hui Y, Yan H, Zhu H, Qu L, Pan C, Chen H, Lan X. Detection of insertion/deletions (indels) of the <i>ATBF1</i> gene and their effects on growth-related traits in three indigenous goat breeds. Arch Anim Breed 2018. [DOI: 10.5194/aab-61-311-2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. The AT motif-binding factor (ATBF1), also known as zinc finger homeobox 3
(ZFXH3), is necessary for activating the POU1F1 gene; thus,
the ATBF1 gene greatly affects the grow traits in animals. The
objective of this work was to explore novel indel (insertion/deletion)
variations and their associations with growth traits in three native Chinese
goat breeds. Two indels within the ATBF1 gene were found in the
Shaanbei white cashmere goat (SWCG; n=581), the Guanzhong dairy goat
(GZDG; n=334) and the Hainan black goat (HNBG; n=270) for the first time
using 12 pairs of primers. Association analysis revealed that the P1-12-bp
indel was consistently correlated with the body height of the three breeds,
and individuals with ID (insertion/deletion) and
DD (deletion/deletion) genotypes had a higher body weight than the
II (insertion/insertion) genotype (P=0.036); the P11-6-bp indel
was consistently correlated with chest circumference and hip width of the
three breeds. Moreover, these two loci were associated with other several
growth-related traits in different breeds. Hence, these findings indicated
that the goat ATBF1 gene had marked effects on growth traits and the
growth-trait-related loci, which would contribute to improving the
growth-related traits of local breeds in the goat industry by implementing
marker-assisted selection (MAS).
Collapse
|
6
|
Xu H, Zhang S, Zhang X, Dang R, Lei C, Chen H, Lan X. Evaluation of novel SNPs and haplotypes within the <i>ATBF1</i> gene and their effects on economically important production traits in cattle. Arch Anim Breed 2017. [DOI: 10.5194/aab-60-285-2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. AT motif binding factor 1 (ATBF1) gene can promote the expression level of the growth hormone 1 (GH1) gene by binding to the enhancers of the POU1F1 and PROP1 genes; thus, it affects the growth and development of livestock. Considering that the ATBF1 gene also has a close relationship with the Janus kinase–signal transductor and activator of transcription (JAK–STAT) pathway, the objective of this work was to identify novel single-nucleotide polymorphism (SNP) variations and their association with growth traits in native Chinese cattle breeds. Five novel SNPs within the ATBF1 gene were found in 644 Qinchuan and Jinnan cattle for first time using 25 pairs of screening and genotyping primers. The five novel SNPs were named as AC_000175:g.140344C>G (SNP1), g.146573T>C (SNP2), g.205468C>T (SNP3), g.205575A>G (SNP4) and g.297690C<T (SNP5). Among them, SNP1 and SNP2 were synonymous coding SNPs, while SNP5 was a missense coding SNP, and the other SNPs were intronic. Haplotype analysis found 18 haplotypes in the two breeds, and three and five closely linked loci were revealed in Qinchuan and Jinnan breeds, respectively. Association analysis revealed that SNP1 was significantly associated with the height across the hip in Qinchuan cattle. SNP2 was found to be significantly related to chest circumference and body side length traits in Jinnan cattle. SNP3 was found to have significant associations with four growth traits in Qinchuan cattle. Moreover, the different combined genotypes, SNP1–SNP3, SNP1–SNP4 and SNP2–SNP5 were significantly associated with the growth traits in cattle. These findings indicated that the bovine ATBF1 gene had marked effects on growth traits, and the growth-trait-related loci can be used as DNA markers for maker-assisted selection (MAS) breeding programs in cattle.
Collapse
|
7
|
Li M, Zhang C, Zhong Y, Zhao J. Cellular localization of ATBF1 protein and its functional implication in breast epithelial cells. Biochem Biophys Res Commun 2017. [DOI: 10.1016/j.bbrc.2017.06.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
8
|
Mortensen AH, Camper SA. Cocaine-and Amphetamine Regulated Transcript (CART) Peptide Is Expressed in Precursor Cells and Somatotropes of the Mouse Pituitary Gland. PLoS One 2016; 11:e0160068. [PMID: 27685990 PMCID: PMC5042496 DOI: 10.1371/journal.pone.0160068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Cocaine-and Amphetamine Regulated Transcript (CART) peptide is expressed in the brain, endocrine and neuroendocrine systems and secreted into the serum. It is thought to play a role in regulation of hypothalamic pituitary functions. Here we report a spatial and temporal analysis of Cart expression in the pituitaries of adult and developing normal and mutant mice with hypopituitarism. We found that Prop1 is not necessary for initiation of Cart expression in the fetal pituitary at e14.5, but it is required indirectly for maintenance of Cart expression in the postnatal anterior pituitary gland. Pou1f1 deficiency has no effect on Cart expression before or after birth. There is no 1:1 correspondence between CART and any particular cell type. In neonates, CART is detected primarily in non-proliferating, POU1F1-positive cells. CART is also found in some cells that express TSH and GH suggesting a correspondence with committed progenitors of the POU1F1 lineage. In summary, we have characterized the normal temporal and cell specific expression of CART in mouse development and demonstrate that postnatal CART expression in the pituitary gland requires PROP1.
Collapse
Affiliation(s)
- Amanda H. Mortensen
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109–5618, United States of America
| | - Sally A. Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48109–5618, United States of America
- * E-mail:
| |
Collapse
|
9
|
Parsons MJ, Brancaccio M, Sethi S, Maywood ES, Satija R, Edwards JK, Jagannath A, Couch Y, Finelli MJ, Smyllie NJ, Esapa C, Butler R, Barnard AR, Chesham JE, Saito S, Joynson G, Wells S, Foster RG, Oliver PL, Simon MM, Mallon AM, Hastings MH, Nolan PM. The Regulatory Factor ZFHX3 Modifies Circadian Function in SCN via an AT Motif-Driven Axis. Cell 2015; 162:607-21. [PMID: 26232227 PMCID: PMC4537516 DOI: 10.1016/j.cell.2015.06.060] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 03/25/2015] [Accepted: 06/01/2015] [Indexed: 01/17/2023]
Abstract
We identified a dominant missense mutation in the SCN transcription factor Zfhx3, termed short circuit (Zfhx3(Sci)), which accelerates circadian locomotor rhythms in mice. ZFHX3 regulates transcription via direct interaction with predicted AT motifs in target genes. The mutant protein has a decreased ability to activate consensus AT motifs in vitro. Using RNA sequencing, we found minimal effects on core clock genes in Zfhx3(Sci/+) SCN, whereas the expression of neuropeptides critical for SCN intercellular signaling was significantly disturbed. Moreover, mutant ZFHX3 had a decreased ability to activate AT motifs in the promoters of these neuropeptide genes. Lentiviral transduction of SCN slices showed that the ZFHX3-mediated activation of AT motifs is circadian, with decreased amplitude and robustness of these oscillations in Zfhx3(Sci/+) SCN slices. In conclusion, by cloning Zfhx3(Sci), we have uncovered a circadian transcriptional axis that determines the period and robustness of behavioral and SCN molecular rhythms.
Collapse
Affiliation(s)
- Michael J Parsons
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Marco Brancaccio
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Siddharth Sethi
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Elizabeth S Maywood
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Rahul Satija
- New York Genome Center, 101 Avenue of the Americas, New York, NY 10013, USA; Department of Biology, New York University, New York, NY 10012, USA
| | - Jessica K Edwards
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Aarti Jagannath
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yvonne Couch
- Acute Stroke Program, Radcliffe Department of Clinical Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Mattéa J Finelli
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Nicola J Smyllie
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Christopher Esapa
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Rachel Butler
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Alun R Barnard
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Johanna E Chesham
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Shoko Saito
- Department of Genetics, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands; Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Greg Joynson
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Sara Wells
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Russell G Foster
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Peter L Oliver
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Michelle M Simon
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Ann-Marie Mallon
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Patrick M Nolan
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, UK.
| |
Collapse
|
10
|
Zhang X, Wu X, Jia W, Pan C, Li X, Lei C, Chen H, Lan X. Novel Nucleotide Variations, Haplotypes Structure and Associations with Growth Related Traits of Goat AT Motif-Binding Factor (ATBF1) Gene. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2015; 28:1394-406. [PMID: 26323396 PMCID: PMC4554846 DOI: 10.5713/ajas.14.0860] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/11/2015] [Accepted: 04/01/2015] [Indexed: 01/05/2023]
Abstract
The AT motif-binding factor (ATBF1) not only interacts with protein inhibitor of activated signal transducer and activator of transcription 3 (STAT3) (PIAS3) to suppress STAT3 signaling regulating embryo early development and cell differentiation, but is required for early activation of the pituitary specific transcription factor 1 (Pit1) gene (also known as POU1F1) critically affecting mammalian growth and development. The goal of this study was to detect novel nucleotide variations and haplotypes structure of the ATBF1 gene, as well as to test their associations with growth-related traits in goats. Herein, a total of seven novel single nucleotide polymorphisms (SNPs) (SNP 1-7) within this gene were found in two well-known Chinese native goat breeds. Haplotypes structure analysis demonstrated that there were four haplotypes in Hainan black goat while seventeen haplotypes in Xinong Saanen dairy goat, and both breeds only shared one haplotype (hap1). Association testing revealed that the SNP2, SNP5, SNP6, and SNP7 loci were also found to significantly associate with growth-related traits in goats, respectively. Moreover, one diplotype in Xinong Saanen dairy goats significantly linked to growth related traits. These preliminary findings not only would extend the spectrum of genetic variations of the goat ATBF1 gene, but also would contribute to implementing marker-assisted selection in genetics and breeding in goats.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xianfeng Wu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wenchao Jia
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chuanying Pan
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiangcheng Li
- Institute of Beijing Animal Science and Veterinary, Chinese Academy of Agricultural Science, Beijing 100194, China
| | - Chuzhao Lei
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hong Chen
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xianyong Lan
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
11
|
Zhai C, Cong H, Liu Y, Zhang Y, Liu X, Zhang H, Ren Z. Rs7193343 polymorphism in zinc finger homeobox 3 (ZFHX3) gene and atrial fibrillation: an updated meta-analysis of 10 case-control comparisons. BMC Cardiovasc Disord 2015; 15:58. [PMID: 26112950 PMCID: PMC4480884 DOI: 10.1186/s12872-015-0044-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 06/01/2015] [Indexed: 01/08/2023] Open
Abstract
Background The previous genome-wide studies have shown that rs7193343 single-nucleotide polymorphism (SNP) in zinc finger homeobox 3 (ZFHX3) gene correlate with risk of atrial fibrillation (AF). However, the distribution of this SNP differs significantly among various populations. The present study was to investigate whether combined evidence shows the association between ZFHX3 rs7193343 SNP and the risk of AF in various populations. Methods A systematic search of all studies published through Dec 2014 was conducted using the Medline, Embase, WanFang, ScienceDirect, CNKI, and OVID databases. The case-control studies that evaluated an association between rs7193343 SNP and risk of AF were identified. The association between the ZFHX3 rs7193343 SNP and AF susceptibility was assessed using genetic models. Results We collected 10 comparisons from six studies for rs7193343 SNP, including 1037 cases and 4310 controls in Asian, 5583 cases and 38215 controls in Caucasian, and then performed an updated meta-analysis and subgroup analysis based on ethnicity. In overall population, the occurrence of AF was found to be associated with T-allelic of rs7193343 SNP in ZFHX3 (OR =1.17, 95% CI 1.10-1.26). In subgroup analysis, we observed there was significant association between T-allele of rs7193343 and risk of AF in Caucasian subgroups (OR =1.20, 95% CI 1.12-1.30), but no statistically significance (OR = 1.07, 95% CI 0.92-1.24) in Asian population. Conclusion In Caucasian population, genetic variant rs7193343 SNP is associated with risk of AF in Caucasian population. However, no association is found in Asian population based on the current evidence. Further studies with larger sample size involving case-control populations with multiple ethnics are still required in the future.
Collapse
Affiliation(s)
- ChuanNan Zhai
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China. .,Graduate School, Tianjin Medical University, Tianjin, 300051, China. .,Department of Cardiology, Tianjin Gongan Hospital, Xinhua Road No. 162, Heping District, Tianjin, 300042, China.
| | - HongLiang Cong
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China.
| | - YuJie Liu
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China.
| | - Ying Zhang
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China.
| | - XianFeng Liu
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China. .,Graduate School, Tianjin Medical University, Tianjin, 300051, China.
| | - Hao Zhang
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China. .,Graduate School, Tianjin Medical University, Tianjin, 300051, China.
| | - ZhiJing Ren
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang South Road No. 291, Jinnan District, Tianjin, 300350, China. .,Graduate School, Tianjin Medical University, Tianjin, 300051, China.
| |
Collapse
|
12
|
An autoregulatory pathway establishes the definitive chromatin conformation at the pit-1 locus. Mol Cell Biol 2015; 35:1523-32. [PMID: 25691665 DOI: 10.1128/mcb.01283-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/09/2015] [Indexed: 12/12/2022] Open
Abstract
The transcription factor Pit-1 (POU1-F1) plays a dominant role in cell lineage expansion and differentiation in the anterior pituitary. Prior studies of the mouse Pit-1 (mPit-1) gene revealed that this master regulatory locus is activated at embryonic day 13.5 (E13.5) by an early enhancer (EE), whereas its subsequent expression throughout adult life is maintained by a more distal definitive enhancer (DE). Here, we demonstrate that the sequential actions of these two enhancers are linked to corresponding shifts in their proximities to the Pit-1 promoter. We further demonstrate that the looping of the definitive enhancer to the mPit-1 promoter is critically dependent on a self-sustaining autoregulatory mechanism mediated by the Pit-1 protein. These Pit-1-dependent actions are accompanied by localized recruitment of CBP and enrichment for H3K27 acetylation within the Pit-1 locus. These data support a model in which the sequential actions of two developmentally activated enhancers are linked to a corresponding shift in higher-order chromatin structures. This shift establishes an autoregulatory circuit that maintains durable expression of Pit-1 throughout adult life.
Collapse
|
13
|
Sun X, Li J, Dong FN, Dong JT. Characterization of nuclear localization and SUMOylation of the ATBF1 transcription factor in epithelial cells. PLoS One 2014; 9:e92746. [PMID: 24651376 PMCID: PMC3961433 DOI: 10.1371/journal.pone.0092746] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/24/2014] [Indexed: 11/30/2022] Open
Abstract
ATBF1/ZFHX3 is a large transcription factor that functions in development, tumorigenesis and other biological processes. ATBF1 is normally localized in the nucleus, but is often mislocalized in the cytoplasm in cancer cells. The mechanism underlying the mislocalization of ATBF1 is unknown. In this study, we analyzed the nuclear localization of ATBF1, and found that ectopically expressed ATBF1 formed nuclear body (NB)-like dots in the nucleus, some of which indeed physically associated with promyelocytic leukemia (PML) NBs. We also defined a 3-amino acid motif, KRK2615-2617, as the nuclear localization signal (NLS) for ATBF1. Interestingly, diffusely distributed nuclear SUMO1 proteins were sequestered into ATBF1 dots, which could be related to ATBF1's physical association with PML NBs, known SUMOylation hotspots. Furthermore, ATBF1 itself was SUMOylated. ATBF1 SUMOylation occurred at more than 3 lysine residues including K2349, K2806 and K3258 and was nuclear specific. Finally, the PIAS3 SUMO1 E3 ligase, which interacts with ATBF1 directly, diminished rather than enhanced ATBF1 SUMOylation, preventing the co-localization of ATBF1 with SUMO1 in the nucleus. These findings suggest that nuclear localization and SUMOylation are important for the transcription factor function of ATBF1, and that ATBF1 could cooperate with PML NBs to regulate protein SUMOylation in different biological processes.
Collapse
Affiliation(s)
- Xiaodong Sun
- Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jie Li
- Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Frederick N. Dong
- Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jin-Tang Dong
- Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
14
|
Tao Y, Zhang M, Li L, Bai Y, Zhou Y, Moon AM, Kaminski HJ, Martin JF. Pitx2, an atrial fibrillation predisposition gene, directly regulates ion transport and intercalated disc genes. CIRCULATION. CARDIOVASCULAR GENETICS 2014; 7:23-32. [PMID: 24395921 PMCID: PMC4013500 DOI: 10.1161/circgenetics.113.000259] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Pitx2 is the homeobox gene located in proximity to the human 4q25 familial atrial fibrillation (AF) locus. When deleted in the mouse germline, Pitx2 haploinsufficiency predisposes to pacing-induced AF, indicating that reduced Pitx2 promotes an arrhythmogenic substrate. Previous work focused on Pitx2 developmental functions that predispose to AF. Although Pitx2 is expressed in postnatal left atrium, it is unknown whether Pitx2 has distinct postnatal and developmental functions. METHODS AND RESULTS To investigate Pitx2 postnatal function, we conditionally inactivated Pitx2 in the postnatal atrium while leaving its developmental function intact. Unstressed adult Pitx2 homozygous mutant mice display variable R-R interval with diminished P-wave amplitude characteristic of sinus node dysfunction, an AF risk factor in human patients. An integrated genomics approach in the adult heart revealed Pitx2 target genes encoding cell junction proteins, ion channels, and critical transcriptional regulators. Importantly, many Pitx2 target genes have been implicated in human AF by genome-wide association studies. Immunofluorescence and transmission electron microscopy studies in adult Pitx2 mutant mice revealed structural remodeling of the intercalated disc characteristic of human patients with AF. CONCLUSIONS Our findings, revealing that Pitx2 has genetically separable postnatal and developmental functions, unveil direct Pitx2 target genes that include channel and calcium handling genes, as well as genes that stabilize the intercalated disc in postnatal atrium.
Collapse
Affiliation(s)
- Ye Tao
- Department of Molecular Physiology and Biophysics, Texas Heart Institute, Houston, TX
| | - Min Zhang
- Department of Molecular Physiology and Biophysics, Texas Heart Institute, Houston, TX
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX
| | - Lele Li
- Department of Molecular Physiology and Biophysics, Texas Heart Institute, Houston, TX
- Cardiomyocyte Renewal Lab, Texas Heart Institute, Houston, TX
| | - Yan Bai
- Department of Molecular Physiology and Biophysics, Texas Heart Institute, Houston, TX
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX
| | - Yuefang Zhou
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Anne M. Moon
- Weis Center for Research, Geisinger Clinic, Danville PA
| | - Henry J. Kaminski
- Department of Neurology, George Washington University, Washington, DC
| | - James F. Martin
- Department of Molecular Physiology and Biophysics, Texas Heart Institute, Houston, TX
- Cardiomyocyte Renewal Lab, Texas Heart Institute, Houston, TX
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX
- Program in Developmental Biology, Baylor College of Medicine, Texas Heart Institute, Houston, TX
| |
Collapse
|
15
|
Tacheny A, Dieu M, Arnould T, Renard P. Mass spectrometry-based identification of proteins interacting with nucleic acids. J Proteomics 2013; 94:89-109. [PMID: 24060998 DOI: 10.1016/j.jprot.2013.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 08/19/2013] [Accepted: 09/13/2013] [Indexed: 01/02/2023]
Abstract
The identification of the regulatory proteins that control DNA transcription as well as RNA stability and translation represents a key step in the comprehension of gene expression regulation. Those proteins can be purified by DNA- or RNA-affinity chromatography, followed by identification by mass spectrometry. Although very simple in the concept, this represents a real technological challenge due to the low abundance of regulatory proteins compared to the highly abundant proteins binding to nucleic acids in a nonsequence-specific manner. Here we review the different strategies that have been set up to reach this purpose, discussing the key parameters that should be considered to increase the chances of success. Typically, two categories of biological questions can be distinguished: the identification of proteins that specifically interact with a precisely defined binding site, mostly addressed by quantitative mass spectrometry, and the identification in a non-comparative manner of the protein complexes recruited by a poorly characterized long regulatory region of nucleic acids. Finally, beside the numerous studies devoted to in vitro-assembled nucleic acid-protein complexes, the scarce data reported on proteomic analyses of in vivo-assembled complexes are described, with a special emphasis on the associated challenges.
Collapse
Affiliation(s)
- A Tacheny
- Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium
| | | | | | | |
Collapse
|
16
|
Abstract
Atrial fibrillation (AF) is the most-common sustained arrhythmia observed in clinical practice, but response to therapy is highly variable between patients. Current drug therapies to suppress AF are incompletely and unpredictably effective and carry substantial risk of proarrhythmia and noncardiac toxicities. The limited success of therapy for AF is partially the result of heterogeneity of the underlying substrate, interindividual differences in disease mechanisms, and our inability to predict response to therapies in individual patients. In this Review, we discuss the evidence that variability in response to drug therapy is also conditioned by the underlying genetic substrate for AF. Increased susceptibility to AF is mediated through diverse genetic mechanisms, including modulation of the atrial action-potential duration, conduction slowing, and impaired cell-to-cell communication, as well as novel mechanisms, such as regulation of signalling proteins important in the pathogenesis of AF. However, the translation of genetic data to the care of the patients with AF has been limited because of poor understanding of the underlying mechanisms associated with common AF-susceptibility loci, a dearth of prospective, adequately powered studies, and the challenges associated with determining efficacy of antiarrhythmic drugs. What is apparent, however, is the need for appropriately designed, genotype-directed clinical trials.
Collapse
Affiliation(s)
- Dawood Darbar
- Departments of Medicine and Pharmacology, Vanderbilt University School of Medicine, 2215B Garland Avenue, Nashville, TN 37323-6602, USA.
| | | |
Collapse
|
17
|
Distinct chromatin configurations regulate the initiation and the maintenance of hGH gene expression. Mol Cell Biol 2013; 33:1723-34. [PMID: 23428872 DOI: 10.1128/mcb.01166-12] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
For many mammalian genes, initiation of transcription during embryonic development must be subsequently sustained over extensive periods of adult life. It remains unclear whether maintenance of gene expression reflects the same set of pathways as are involved in initial gene activation. The human pituitary growth hormone (hGH-N) locus is activated in the differentiating somatotrope midway through embryogenesis by a multicomponent locus control region (LCR). DNase I-hypersensitive site I (HSI) of the LCR is essential to full developmental activation of the hGH-N locus. Here we demonstrate that conditional deletion of HSI from the active hGH locus in the adult pituitary effectively silences hGH-N expression. Analyses of chromatin structure and locus positioning demonstrate that a specific subset of the HSI functions active in the embryo retain their HSI dependence in the adult pituitary. These functions sustain engagement of the hGH locus with polymerase II (Pol II) factories, histone acetylation at the hGH-N promoter, and looping of the LCR to its target promoter. These data reveal that HSI is essential to both the maintenance and the initiation phases of gene expression. These observations contribute to our mechanistic understanding of how stable patterns of mammalian gene expression are established in a terminally differentiated cell.
Collapse
|
18
|
Li M, Zhao D, Ma G, Zhang B, Fu X, Zhu Z, Fu L, Sun X, Dong JT. Upregulation of ATBF1 by progesterone-PR signaling and its functional implication in mammary epithelial cells. Biochem Biophys Res Commun 2013; 430:358-63. [DOI: 10.1016/j.bbrc.2012.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 11/03/2012] [Indexed: 12/23/2022]
|
19
|
Oh JE, Chambwe N, Klein S, Gal J, Andrews S, Gleason G, Shaknovich R, Melnick A, Campagne F, Toth M. Differential gene body methylation and reduced expression of cell adhesion and neurotransmitter receptor genes in adverse maternal environment. Transl Psychiatry 2013; 3:e218. [PMID: 23340501 PMCID: PMC3566713 DOI: 10.1038/tp.2012.130] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Early life adversity, including adverse gestational and postpartum maternal environment, is a contributing factor in the development of autism, attention deficit hyperactivity disorder (ADHD), anxiety and depression but little is known about the underlying molecular mechanism. In a model of gestational maternal adversity that leads to innate anxiety, increased stress reactivity and impaired vocal communication in the offspring, we asked if a specific DNA methylation signature is associated with the emergence of the behavioral phenotype. Genome-wide DNA methylation analyses identified 2.3% of CpGs as differentially methylated (that is, differentially methylated sites, DMSs) by the adverse environment in ventral-hippocampal granule cells, neurons that can be linked to the anxiety phenotype. DMSs were typically clustered and these clusters were preferentially located at gene bodies. Although CpGs are typically either highly methylated or unmethylated, DMSs had an intermediate (20-80%) methylation level that may contribute to their sensitivity to environmental adversity. The adverse maternal environment resulted in either hyper or hypomethylation at DMSs. Clusters of DMSs were enriched in genes that encode cell adhesion molecules and neurotransmitter receptors; some of which were also downregulated, indicating multiple functional deficits at the synapse in adversity. Pharmacological and genetic evidence links many of these genes to anxiety.
Collapse
Affiliation(s)
- J-e Oh
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA,Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA. E-mail: or
| | - N Chambwe
- Department of Physiology and Biophysics and HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - S Klein
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - J Gal
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - S Andrews
- Department of Physiology and Biophysics and HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - G Gleason
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - R Shaknovich
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - A Melnick
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - F Campagne
- Department of Physiology and Biophysics and HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA
| | - M Toth
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA,Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA. E-mail: or
| |
Collapse
|
20
|
Li M, Fu X, Ma G, Sun X, Dong X, Nagy T, Xing C, Li J, Dong JT. Atbf1 regulates pubertal mammary gland development likely by inhibiting the pro-proliferative function of estrogen-ER signaling. PLoS One 2012; 7:e51283. [PMID: 23251482 PMCID: PMC3520988 DOI: 10.1371/journal.pone.0051283] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 10/31/2012] [Indexed: 11/18/2022] Open
Abstract
ATBF1 is a candidate tumor suppressor that interacts with estrogen receptor (ER) to inhibit the function of estrogen-ER signaling in gene regulation and cell proliferation control in human breast cancer cells. We therefore tested whether Atbf1 and its interaction with ER modulate the development of pubertal mammary gland, where estrogen is the predominant steroid hormone. In an in vitro model of cell differentiation, i.e., MCF10A cells cultured in Matrigel, ATBF1 expression was significantly increased, and knockdown of ATBF1 inhibited acinus formation. During mouse mammary gland development, Atbf1 was expressed at varying levels at different stages, with higher levels during puberty, lower during pregnancy, and the highest during lactation. Knockout of Atbf1 at the onset of puberty enhanced ductal elongation and bifurcation and promoted cell proliferation in both ducts and terminal end buds of pubertal mammary glands. Enhanced cell proliferation primarily occurred in ER-positive cells and was accompanied by increased expression of ER target genes. Furthermore, inactivation of Atbf1 reduced the expression of basal cell markers (CK5, CK14 and CD44) but not luminal cell markers. These findings indicate that Atbf1 plays a role in the development of pubertal mammary gland likely by modulating the function of estrogen-ER signaling in luminal cells and by modulating gene expression in basal cells.
Collapse
Affiliation(s)
- Mei Li
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Xiaoying Fu
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Gui Ma
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaodong Sun
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Xueyuan Dong
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (XD) (XD); (JTD) (JD)
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Changsheng Xing
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jie Li
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (XD) (XD); (JTD) (JD)
| |
Collapse
|
21
|
Herriges JC, Yi L, Hines EA, Harvey JF, Xu G, Gray P, Ma Q, Sun X. Genome-scale study of transcription factor expression in the branching mouse lung. Dev Dyn 2012; 241:1432-53. [PMID: 22711520 PMCID: PMC3529173 DOI: 10.1002/dvdy.23823] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Mammalian lung development consists of a series of precisely choreographed events that drive the progression from simple lung buds to the elaborately branched organ that fulfills the vital function of gas exchange. Strict transcriptional control is essential for lung development. Among the large number of transcription factors encoded in the mouse genome, only a small portion of them are known to be expressed and function in the developing lung. Thus a systematic investigation of transcription factors expressed in the lung is warranted. RESULTS To enrich for genes that may be responsible for regional growth and patterning, we performed a screen using RNA in situ hybridization to identify genes that show restricted expression patterns in the embryonic lung. We focused on the pseudoglandular stage during which the lung undergoes branching morphogenesis, a cardinal event of lung development. Using a genome-scale probe set that represents over 90% of the transcription factors encoded in the mouse genome, we identified 62 transcription factor genes with localized expression in the epithelium, mesenchyme, or both. Many of these genes have not been previously implicated in lung development. CONCLUSIONS Our findings provide new starting points for the elucidation of the transcriptional circuitry that controls lung development.
Collapse
Affiliation(s)
- John C. Herriges
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Lan Yi
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Elizabeth A. Hines
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Julie F. Harvey
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| | - Guoliang Xu
- Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China 200031
| | - Paul Gray
- Department of Anatomy and Neurobiology, Washington University, St. Louis, MO 63110
| | - Qiufu Ma
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Xin Sun
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
22
|
Tacheny A, Michel S, Dieu M, Payen L, Arnould T, Renard P. Unbiased proteomic analysis of proteins interacting with the HIV-1 5'LTR sequence: role of the transcription factor Meis. Nucleic Acids Res 2012; 40:e168. [PMID: 22904091 PMCID: PMC3505963 DOI: 10.1093/nar/gks733] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To depict the largest picture of a core promoter interactome, we developed a one-step DNA-affinity capture method coupled with an improved mass spectrometry analysis process focused on the identification of low abundance proteins. As a proof of concept, this method was developed through the analysis of 230 bp contained in the 5′long terminal repeat (LTR) of the human immunodeficiency virus 1 (HIV-1). Beside many expected interactions, many new transcriptional regulators were identified, either transcription factors (TFs) or co-regulators, which interact directly or indirectly with the HIV-1 5′LTR. Among them, the homeodomain-containing TF myeloid ectopic viral integration site was confirmed to functionally interact with a specific binding site in the HIV-1 5′LTR and to act as a transcriptional repressor, probably through recruitment of the repressive Sin3A complex. This powerful and validated DNA-affinity approach could also be used as an efficient screening tool to identify a large set of proteins that physically interact, directly or indirectly, with a DNA sequence of interest. Combined with an in silico analysis of the DNA sequence of interest, this approach provides a powerful approach to select the interacting candidates to validate functionally by classical approaches.
Collapse
Affiliation(s)
- A Tacheny
- Laboratory of Biochemistry and Cell Biology (URBC), NAmur Research Institute for LIfe Sciences, University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium
| | | | | | | | | | | |
Collapse
|
23
|
Nicholas TJ, Baker C, Eichler EE, Akey JM. A high-resolution integrated map of copy number polymorphisms within and between breeds of the modern domesticated dog. BMC Genomics 2011; 12:414. [PMID: 21846351 PMCID: PMC3166287 DOI: 10.1186/1471-2164-12-414] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 08/16/2011] [Indexed: 01/22/2023] Open
Abstract
Background Structural variation contributes to the rich genetic and phenotypic diversity of the modern domestic dog, Canis lupus familiaris, although compared to other organisms, catalogs of canine copy number variants (CNVs) are poorly defined. To this end, we developed a customized high-density tiling array across the canine genome and used it to discover CNVs in nine genetically diverse dogs and a gray wolf. Results In total, we identified 403 CNVs that overlap 401 genes, which are enriched for defense/immunity, oxidoreductase, protease, receptor, signaling molecule and transporter genes. Furthermore, we performed detailed comparisons between CNVs located within versus outside of segmental duplications (SDs) and find that CNVs in SDs are enriched for gene content and complexity. Finally, we compiled all known dog CNV regions and genotyped them with a custom aCGH chip in 61 dogs from 12 diverse breeds. These data allowed us to perform the first population genetics analysis of canine structural variation and identify CNVs that potentially contribute to breed specific traits. Conclusions Our comprehensive analysis of canine CNVs will be an important resource in genetically dissecting canine phenotypic and behavioral variation.
Collapse
Affiliation(s)
- Thomas J Nicholas
- Department of Genome Sciences, University of Washington, 1705 NE Pacific, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
24
|
Angotzi AR, Mungpakdee S, Stefansson S, Male R, Chourrout D. Involvement of Prop1 homeobox gene in the early development of fish pituitary gland. Gen Comp Endocrinol 2011; 171:332-40. [PMID: 21362424 DOI: 10.1016/j.ygcen.2011.02.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/17/2011] [Accepted: 02/21/2011] [Indexed: 11/22/2022]
Abstract
When mutated in mammals, paired-like homeobox Prop1 gene produces highly variable pituitary phenotypes with impaired regulation of Pit1 and eventually defective synthesis of Pit1-regulated pituitary hormones. Here we have identified fish prop1 orthologs, confirmed their pituitary-specific expression, and blocked the splicing of zebrafish prop1 transcripts using morpholino oligonucleotides. Very early steps of the gland formation seemed unaffected based on morphology and expression of early placodal marker pitx. Prop1 knock-down reduced the expression of pit1, prl (prolactin) and gh (growth hormone), as expected if the function of Prop1 is conserved throughout vertebrates. Less expectedly, lim3 was down regulated. This gene is expressed from early stages of vertebrate pituitary development but is not known to be Prop1-dependent. In situ hybridizations on prop1 morphants using probes for the pan pituitary gene pitx3 and for the hormone gene markers prl, gh and tshβ, revealed abnormal shape, growth and cellular organization of the developed adenohypophysis. Strikingly, the effects of prop1 knock-down on adenohypophysis morphology and gene expression were gradually reversed during late development, despite persistent splice-blocking of transcripts. Therefore, prop1 function appears to be conserved between mammals and fish, at least for the mediation of hormonal cell type differentiation via pit1, but the existence of other fish-specific pathways downstream of prop1 are suggested by our observations.
Collapse
Affiliation(s)
- Anna Rita Angotzi
- Sars, International Centre for Marine Molecular Biology, University of Bergen, High Technology Centre, Thormoehlensgt 55, N-5008 Bergen, Norway
| | | | | | | | | |
Collapse
|
25
|
Guillaumond F, Boyer B, Becquet D, Guillen S, Kuhn L, Garin J, Belghazi M, Bosler O, Franc J, François‐Bellan A. Chromatin remodeling as a mechanism for circadian prolactin transcription: rhythmic NONO and SFPQ recruitment to HLTF. FASEB J 2011; 25:2740-56. [DOI: 10.1096/fj.10-178616] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Fabienne Guillaumond
- Institut des Sciences Moleculaires de Marseille (ISM2)UMR6263 Université Aix‐Marseille IIIMarseilleFrance
| | - Benedicte Boyer
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6231Université Aix‐Marseille II, IIIMarseilleFrance
| | - Denis Becquet
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6231Université Aix‐Marseille II, IIIMarseilleFrance
| | - Severine Guillen
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6231Université Aix‐Marseille II, IIIMarseilleFrance
| | - Lauriane Kuhn
- Plateforme Étude de la Dynamique des Protéomes (EDyP)‐ServiceGrenobleFrance
| | - Jerome Garin
- Centre d'Analyse Protéomique de MarseilleInstitut Fédératif de Recherche (IFR) Jean‐RocheMarseilleFrance
| | - Maya Belghazi
- Plateforme Protéomique de l'Esplanade Institut de Biologie Moléculaire et Cellulaire (IBMC)StrasbourgFrance
| | - Olivier Bosler
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6231Université Aix‐Marseille II, IIIMarseilleFrance
| | - Jean‐Louis Franc
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6231Université Aix‐Marseille II, IIIMarseilleFrance
| | - Anne‐Marie François‐Bellan
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M)Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 6231Université Aix‐Marseille II, IIIMarseilleFrance
| |
Collapse
|
26
|
Kelberman D, Rizzoti K, Lovell-Badge R, Robinson ICAF, Dattani MT. Genetic regulation of pituitary gland development in human and mouse. Endocr Rev 2009; 30:790-829. [PMID: 19837867 PMCID: PMC2806371 DOI: 10.1210/er.2009-0008] [Citation(s) in RCA: 278] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Normal hypothalamopituitary development is closely related to that of the forebrain and is dependent upon a complex genetic cascade of transcription factors and signaling molecules that may be either intrinsic or extrinsic to the developing Rathke's pouch. These factors dictate organ commitment, cell differentiation, and cell proliferation within the anterior pituitary. Abnormalities in these processes are associated with congenital hypopituitarism, a spectrum of disorders that includes syndromic disorders such as septo-optic dysplasia, combined pituitary hormone deficiencies, and isolated hormone deficiencies, of which the commonest is GH deficiency. The highly variable clinical phenotypes can now in part be explained due to research performed over the last 20 yr, based mainly on naturally occurring and transgenic animal models. Mutations in genes encoding both signaling molecules and transcription factors have been implicated in the etiology of hypopituitarism, with or without other syndromic features, in mice and humans. To date, mutations in known genes account for a small proportion of cases of hypopituitarism in humans. However, these mutations have led to a greater understanding of the genetic interactions that lead to normal pituitary development. This review attempts to describe the complexity of pituitary development in the rodent, with particular emphasis on those factors that, when mutated, are associated with hypopituitarism in humans.
Collapse
Affiliation(s)
- Daniel Kelberman
- Developmental Endocrinology Research Group, Clinical and Molecular Genetics Unit, Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | | | | | | | | |
Collapse
|
27
|
Gudbjartsson DF, Holm H, Gretarsdottir S, Thorleifsson G, Walters GB, Thorgeirsson G, Gulcher J, Mathiesen E, Njølstad I, Nyrnes A, Wilsgaard T, Hald E, Hveem K, Stoltenberg C, Kucera G, Stubblefield T, Carter S, Roden D, Ng MC, Baum L, So WY, Wong KS, Chan JCN, Gieger C, Wichmann HE, Gschwendtner A, Dichgans M, Kuhlenbäumer G, Berger K, Ringelstein EB, Bevan S, Markus H, Kostulas K, Hillert J, Sveinbjörnsdóttir S, Valdimarsson EM, Løchen ML, Ma RCW, Darbar D, Kong A, Arnar DO, Thorsteinsdottir U, Stefansson K. A sequence variant in ZFHX3 on 16q22 associates with atrial fibrillation and ischemic stroke. Nat Genet 2009; 41:876-8. [PMID: 19597491 PMCID: PMC2740741 DOI: 10.1038/ng.417] [Citation(s) in RCA: 370] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Accepted: 06/11/2009] [Indexed: 11/09/2022]
Abstract
We expanded our genome-wide association study on atrial fibrillation (AF) in Iceland, which previously identified risk variants on 4q25, and tested the most significant associations in samples from Iceland, Norway and the United States. A variant in the ZFHX3 gene on chromosome 16q22, rs7193343-T, associated significantly with AF (odds ratio OR = 1.21, P = 1.4 x 10(-10)). This variant also associated with ischemic stroke (OR = 1.11, P = 0.00054) and cardioembolic stroke (OR = 1.22, P = 0.00021) in a combined analysis of five stroke samples.
Collapse
Affiliation(s)
| | - Hilma Holm
- DeCODE genetics, Sturlugata 8, 101 Reykjavik, Iceland
- Department of Internal Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | - Gudmundur Thorgeirsson
- Department of Internal Medicine, Division of Cardiology, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Ellisiv Mathiesen
- Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
| | - Inger Njølstad
- Institute of Community Medicine, University of Tromsø, Tromsø, Norway
| | - Audhild Nyrnes
- Institute of Community Medicine, University of Tromsø, Tromsø, Norway
- Department of Geriatrics, University Hospital of North Norway, Tromsø, Norway
| | - Tom Wilsgaard
- Institute of Community Medicine, University of Tromsø, Tromsø, Norway
| | - Erin Hald
- Department of Cardiology, University Hospital of North Norway, Tromsø, Norway
| | - Kristian Hveem
- Department of Public Health and General Practice, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Gayle Kucera
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Tanya Stubblefield
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Shannon Carter
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Dan Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | - Maggie C.Y. Ng
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Larry Baum
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Wing Yee So
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Ka Sing Wong
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Juliana C. N. Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - H-Erich Wichmann
- Institute of Epidemiology, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Andreas Gschwendtner
- Department of Neurology, Klinikum of Grosshadern, Ludwig-Maximilians-University Munich, München, Germany
| | - Martin Dichgans
- Department of Neurology, Klinikum of Grosshadern, Ludwig-Maximilians-University Munich, München, Germany
| | - Gregor Kuhlenbäumer
- Institute of Experimental Medicine, Molecular Neurobiology, University of Kiel, Kiel, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Muenster, Muenster, Germany
| | - E. Bernd Ringelstein
- Department of Neurology and Leibniz-Institute for Atherosclerosis Research, University of Muenster, Muenster, Germany
| | - Steve Bevan
- Department of Clinical Neuroscience, St. George's University of London, London, United Kingdom
| | - Hugh Markus
- Department of Clinical Neuroscience, St. George's University of London, London, United Kingdom
| | | | - Jan Hillert
- Department of Neurology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | - Maja-Lisa Løchen
- Institute of Community Medicine, University of Tromsø, Tromsø, Norway
- Department of Cardiology, University Hospital of North Norway, Tromsø, Norway
| | - Ronald C. W. Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Dawood Darbar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennesse, USA
| | | | - David O. Arnar
- Department of Internal Medicine, Division of Cardiology, Landspitali University Hospital, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- DeCODE genetics, Sturlugata 8, 101 Reykjavik, Iceland
- University of Iceland, Faculty of Medicine, 101 Reykjavik
| | - Kari Stefansson
- DeCODE genetics, Sturlugata 8, 101 Reykjavik, Iceland
- University of Iceland, Faculty of Medicine, 101 Reykjavik
| |
Collapse
|
28
|
Davis SW, Potok MA, Brinkmeier ML, Carninci P, Lyons RH, MacDonald JW, Fleming MT, Mortensen AH, Egashira N, Ghosh D, Steel KP, Osamura RY, Hayashizaki Y, Camper SA. Genetics, gene expression and bioinformatics of the pituitary gland. HORMONE RESEARCH 2009; 71 Suppl 2:101-15. [PMID: 19407506 PMCID: PMC3140954 DOI: 10.1159/000192447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetic cases of congenital pituitary hormone deficiency are common and many are caused by transcription factor defects. Mouse models with orthologous mutations are invaluable for uncovering the molecular mechanisms that lead to problems in organ development and typical patient characteristics. We are using mutant mice defective in the transcription factors PROP1 and POU1F1 for gene expression profiling to identify target genes for these critical transcription factors and candidates for cases of pituitary hormone deficiency of unknown aetiology. These studies reveal critical roles for Wnt signalling pathways, including the TCF/LEF transcription factors and interacting proteins of the groucho family, bone morphogenetic protein antagonists and targets of notch signalling. Current studies are investigating the roles of novel homeobox genes and pathways that regulate the transition from proliferation to differentiation, cell adhesion and cell migration. Pituitary adenomas are a common human health problem, yet most cases are sporadic, necessitating alternative approaches to traditional Mendelian genetic studies. Mouse models of adenoma formation offer the opportunity for gene expression profiling during progressive stages of hyperplasia, adenoma and tumorigenesis. This approach holds promise for the identification of relevant pathways and candidate genes as risk factors for adenoma formation, understanding mechanisms of progression, and identifying drug targets and clinically relevant biomarkers.
Collapse
Affiliation(s)
| | | | | | - Piero Carninci
- Omics Science Center, RIKEN Yokohama Institute, Yokohama
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kerr J, Wood W, Ridgway EC. Basic science and clinical research advances in the pituitary transcription factors: Pit-1 and Prop-1. Curr Opin Endocrinol Diabetes Obes 2008; 15:359-63. [PMID: 18594277 DOI: 10.1097/med.0b013e3283060a56] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The pituitary-specific transcription factors, Pit-1 (also called Pou1f1) and prophet of Pit-1 (Prop-1), are critical for normal pituitary development and function. The aim of the present review is to highlight the recent basic science and clinical research advances in these transcription factors, as they relate to signaling pathway interactions, gene regulation, and mutations in human diseases. RECENT FINDINGS A number of important basic research discoveries have been made in the area of the pituitary-specific transcription factors, Pit-1 and Prop-1. Among these findings include: the effects of the Pit-1 coactivators, GATA-2 and TRAP-220, on the transcriptional regulation of the TSHbeta gene and thyrotropin expression, characterization of a novel pituitary regulator of Pit-1 expression, Atbf1, elucidation of the roles of Wnt and Notch signaling on Prop-1-mediated specification of the Pit-1 cell lineage and gonadotropes, and the identification of regulatory regions of the Prop-1 gene. Advances in clinical research include: the identification of novel mutations in the human POU1F1 and PROP-1 genes, and screening guidelines for patients with combined pituitary hormone deficiencies and possible mutations in these transcription factors. SUMMARY Research into the complex interplay of signaling pathways and transcription factors that regulate the pituitary gland are important areas of developmental biology and normal physiology. Clinically, such research has important implications for human diseases by identifying inheritable transcription factor mutations that may cause pituitary hormonal deficiencies.
Collapse
Affiliation(s)
- Janice Kerr
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado, Denver, Aurora, Colorado 80045, USA.
| | | | | |
Collapse
|