1
|
Jiang G, Xue Y, Huang X. Temperature-Induced Sex Differentiation in River Prawn ( Macrobrachium nipponense): Mechanisms and Effects. Int J Mol Sci 2024; 25:1207. [PMID: 38279207 PMCID: PMC10816446 DOI: 10.3390/ijms25021207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
Macrobrachium nipponense is gonochoristic and sexually dimorphic. The male prawn grows faster and usually has a larger size than the female. Therefore, a higher male proportion in stock usually results in higher yield. To investigate the impact of temperature on sexual differentiation in M. nipponense, two temperature treatments (26 °C and 31 °C) were conducted. The results showed that compared to the 31 °C treatment (3.20 ± 0.12), the 26 °C treatment displayed a lower female/male ratio (2.20 ± 0.11), which implied that a lower temperature could induce masculinization in M. nipponense. The temperature-sensitive sex differentiation phase was 25-35 days post hatching (DPH) at 26 °C while 15-20 DPH at 31 °C. Transcriptome and qPCR analysis revealed that a lower temperature up-regulated the expression of genes related to androgen secretion, and down-regulated the expressions of genes related to oogonia differentiation. Thirty-one temperature-regulated sex-differentiation genes were identified and the molecular mechanism of temperature-regulated sex differentiation was suggested. The finding of this study indicates that temperature regulation can be proposed as an innovative strategy for improving the culture yield of M. nipponense.
Collapse
Affiliation(s)
- Gang Jiang
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; (G.J.); (Y.X.)
| | - Yucai Xue
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; (G.J.); (Y.X.)
| | - Xuxiong Huang
- Centre for Research on Environmental Ecology and Fish Nutrition (CREEFN) of the Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China; (G.J.); (Y.X.)
- Building of China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology and Joint Research on Mariculture Technology, Shanghai 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
2
|
Zhang P, Jing K, Tian Y, Li Y, Chai Z, Cai X. Additional glial cell line-derived neurotrophic factor in vitro promotes the proliferation of undifferentiated spermatogonia from sterile cattleyak. Anim Reprod Sci 2024; 260:107385. [PMID: 38056175 DOI: 10.1016/j.anireprosci.2023.107385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/23/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
Cattleyak is a typically male sterile species. The meiosis process is blocked and the scarcity of spermatogenic stems cells are both contributing factors to the inability of male cattleyak to produce sperm. While Glial cell line-derived neurotrophic factor (GDNF) is the first discovered growth factor known to promote the proliferation and self-renewal of spermatogenic stem cells, its relationship to the spermatogenesis arrest of cattleyak remains unclear. In this report, we studied the differential expression of GDNF in the testis of yak and cattleyak, and discussed the optimal concentration of GDNF in the culture medium of undifferentiated spermatogonia (UDSPG) of cattleyak in vitro and the effect of GDNF on the proliferation of cattleyak UDSPG. The results indicated that GDNF expression in the testicular tissue of cattleyak was inferior to that of yak. Moreover, the optimum value for the UDSPG in vitro culture was determined to be 20-30 ng/mL for cattleyak. In vitro, the proliferation activity of UDSPG was observed to increase with additional GDNF due to the up-regulation of proliferation-related genes and the down-regulation of differentiation-related genes. We hereby report that the scarcity of cattleyak UDSPG is due to insufficient expression of GDNF, and that the addition of GDNF in vitro can promote the proliferation of cattleyak UDSPG by regulating the expression of genes related to proliferation and differentiation. This work provides a new insight to solve the issue of spermatogenic arrest in cattleyak.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China
| | - Kemin Jing
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yuan Tian
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yuqian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Dutta Banik D, Martin LJ, Tang T, Soboloff J, Tourtellotte WG, Pierchala BA. EGR4 is critical for cell-fate determination and phenotypic maintenance of geniculate ganglion neurons underlying sweet and umami taste. Proc Natl Acad Sci U S A 2023; 120:e2217595120. [PMID: 37216536 PMCID: PMC10235952 DOI: 10.1073/pnas.2217595120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/23/2023] [Indexed: 05/24/2023] Open
Abstract
The sense of taste starts with activation of receptor cells in taste buds by chemical stimuli which then communicate this signal via innervating oral sensory neurons to the CNS. The cell bodies of oral sensory neurons reside in the geniculate ganglion (GG) and nodose/petrosal/jugular ganglion. The geniculate ganglion contains two main neuronal populations: BRN3A+ somatosensory neurons that innervate the pinna and PHOX2B+ sensory neurons that innervate the oral cavity. While much is known about the different taste bud cell subtypes, considerably less is known about the molecular identities of PHOX2B+ sensory subpopulations. In the GG, as many as 12 different subpopulations have been predicted from electrophysiological studies, while transcriptional identities exist for only 3 to 6. Importantly, the cell fate pathways that diversify PHOX2B+ oral sensory neurons into these subpopulations are unknown. The transcription factor EGR4 was identified as being highly expressed in GG neurons. EGR4 deletion causes GG oral sensory neurons to lose their expression of PHOX2B and other oral sensory genes and up-regulate BRN3A. This is followed by a loss of chemosensory innervation of taste buds, a loss of type II taste cells responsive to bitter, sweet, and umami stimuli, and a concomitant increase in type I glial-like taste bud cells. These deficits culminate in a loss of nerve responses to sweet and umami taste qualities. Taken together, we identify a critical role of EGR4 in cell fate specification and maintenance of subpopulations of GG neurons, which in turn maintain the appropriate sweet and umami taste receptor cells.
Collapse
Affiliation(s)
- Debarghya Dutta Banik
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Louis J. Martin
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Tao Tang
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Jonathan Soboloff
- Department of Cancer & Cellular Biology, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA19140
| | - Warren G. Tourtellotte
- Department of Pathology and Laboratory Medicine, Neurology, and Neurological Surgery, Cedars-Sinai Medical Center, Los Angeles, CA90048
| | - Brian A. Pierchala
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| |
Collapse
|
4
|
Zhang Z, Yang Y, Huang L, Chen L, Zhang G, Gong P, Ye S, Feng Y. Identification of potential candidate genes and regulatory pathways related to reproductive capacity in hypothalamus and pituitarium of male ducks (Anas platyrhynchos) by differential transcriptome analysis. J Anim Sci 2023; 101:skac363. [PMID: 36315611 PMCID: PMC9890447 DOI: 10.1093/jas/skac363] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/29/2022] [Indexed: 11/05/2022] Open
Abstract
The improvement of reproductive capacity of poultry is important for the poultry industry. The existing studies on reproductive capacity mainly focus on the testis tissue, but few reports on regulationary effect of brain neuroendocrime on reproductive capacity have been available. The hypothalamus-pituitarium-gonad (HPG) axis is an important pathway regulating spermatogenesis and sexual behavior. This study analyzed the gene expression in the hypothalamus and pituitary tissues of male ducks in high-semen-quality group (DH), low-semen-quality group (DL), and non-response group (DN) by RNA-sequencing. A total of 1980 differentially expressed genes (DEGs) were identified, and significantly less DEGs were found in pituitary gland than in hypothalamus. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that these DEGs were mainly enriched in nerve-related and synapse-related biological processes, mitochondrial inner membrane formation pathway, and ribosome structure pathway. Notably, the neuroactive ligand-receptor interaction pathway significantly enriched in all three comparisons (DH vs. DL, DH vs. DN, and DL vs. DN) was related to different reproductive performance such as semen quality and sexual response. Furthermore, six genes, including POMC, CPLX2, HAPLN2, EGR4, TOX3, and MSH4, were identified as candidate genes regulating reproductive capacity. Our findings provide new insights into the regulation mechanisms underlying the reproductive performance of male poultry, and offer a valuable reference for duck breeding programs aimed at promoting reproductive capacity.
Collapse
Affiliation(s)
- Zhen Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, 430070 Wuhan, People’s Republic of China
| | - Yu Yang
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, 430208 Wuhan, Hubei, People’s Republic of China
| | - Liming Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, 430070 Wuhan, People’s Republic of China
| | - Ligen Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, 430070 Wuhan, People’s Republic of China
| | - Guixin Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, 430070 Wuhan, People’s Republic of China
| | - Ping Gong
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, 430208 Wuhan, Hubei, People’s Republic of China
| | - Shengqiang Ye
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, 430208 Wuhan, Hubei, People’s Republic of China
| | - Yanping Feng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, 430070 Wuhan, People’s Republic of China
| |
Collapse
|
5
|
Alhasnani MA, Loeb S, Hall SJ, Caruolo Z, Simmonds F, Solano AE, Spade DJ. Interaction between mono-(2-ethylhexyl) phthalate and retinoic acid alters Sertoli cell development during fetal mouse testis cord morphogenesis. Curr Res Toxicol 2022; 3:100087. [PMID: 36189433 PMCID: PMC9520016 DOI: 10.1016/j.crtox.2022.100087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/24/2022] Open
Abstract
Phthalic acid esters (phthalates) are a class of industrial chemicals that cause developmental and reproductive toxicity, but there are significant gaps in knowledge of phthalate toxicity mechanisms. There is evidence that phthalates disrupt retinoic acid signaling in the fetal testis, potentially disrupting control of spatial and temporal patterns of testis development. Our goal was to determine how a phthalate would interact with retinoic acid signaling during fetal mouse testis development. We hypothesized that mono-(2-ethylhexyl) phthalate (MEHP) would exacerbate the adverse effect of all-trans retinoic acid (ATRA) on seminiferous cord development in the mouse fetal testis. To test this hypothesis, gestational day (GD) 14 C57BL/6 mouse testes were isolated and cultured on media containing MEHP, ATRA, or a combination of both compounds. Cultured testes were collected for global transcriptome analysis after one day in culture and for histology and immunofluorescent analysis of Sertoli cell differentiation after three days in culture. ATRA disrupted seminiferous cord morphogenesis and induced aberrant FOXL2 expression. MEHP alone had no significant effect on cord development, but combined exposure to MEHP and ATRA increased the number of FOXL2-positive cells, reduced seminiferous cord number, and increased testosterone levels, beyond the effect of ATRA alone. In RNA-seq analysis, ATRA treatment and MEHP treatment resulted in differential expression of genes 510 and 134 genes, respectively, including 70 common differentially expressed genes (DEGs) between the two treatments, including genes with known roles in fetal testis development. MEHP DEGs included RAR target genes, genes involved in angiogenesis, and developmental patterning genes, including members of the homeobox superfamily. These results support the hypothesis that MEHP modulates retinoic acid signaling in the mouse fetal testis and provide insight into potential mechanisms by which phthalates disrupt seminiferous cord morphogenesis.
Collapse
Key Words
- ATRA, All-trans retinoic acid. CAS # 302-79-4
- DMSO, dimethyl sulfoxide
- Fetal testis development
- GD, gestational day
- GO, Gene Ontology
- IPA, Ingenuity Pathway Analysis
- ITCN, Image-based Tool for Counting Nuclei
- MEHP, mono-(2-ethylheyxl) phthalate. CAS # 4376-20-9
- MNGs, multinucleated germ cells
- PVC, polyvinyl chloride
- Phthalate toxicity
- Retinoic acid
- Sertoli cell
- TDS, testicular dysgenesis syndrome
Collapse
Affiliation(s)
- Maha A. Alhasnani
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Skylar Loeb
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Susan J. Hall
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Zachary Caruolo
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Faith Simmonds
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Amanda E. Solano
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Daniel J. Spade
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| |
Collapse
|
6
|
Liu Y, Kossack ME, McFaul ME, Christensen LN, Siebert S, Wyatt SR, Kamei CN, Horst S, Arroyo N, Drummond IA, Juliano CE, Draper BW. Single-cell transcriptome reveals insights into the development and function of the zebrafish ovary. eLife 2022; 11:e76014. [PMID: 35588359 PMCID: PMC9191896 DOI: 10.7554/elife.76014] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Zebrafish are an established research organism that has made many contributions to our understanding of vertebrate tissue and organ development, yet there are still significant gaps in our understanding of the genes that regulate gonad development, sex, and reproduction. Unlike the development of many organs, such as the brain and heart that form during the first few days of development, zebrafish gonads do not begin to form until the larval stage (≥5 days post-fertilization). Thus, forward genetic screens have identified very few genes required for gonad development. In addition, bulk RNA-sequencing studies that identify genes expressed in the gonads do not have the resolution necessary to define minor cell populations that may play significant roles in the development and function of these organs. To overcome these limitations, we have used single-cell RNA sequencing to determine the transcriptomes of cells isolated from juvenile zebrafish ovaries. This resulted in the profiles of 10,658 germ cells and 14,431 somatic cells. Our germ cell data represents all developmental stages from germline stem cells to early meiotic oocytes. Our somatic cell data represents all known somatic cell types, including follicle cells, theca cells, and ovarian stromal cells. Further analysis revealed an unexpected number of cell subpopulations within these broadly defined cell types. To further define their functional significance, we determined the location of these cell subpopulations within the ovary. Finally, we used gene knockout experiments to determine the roles of foxl2l and wnt9b for oocyte development and sex determination and/or differentiation, respectively. Our results reveal novel insights into zebrafish ovarian development and function, and the transcriptome profiles will provide a valuable resource for future studies.
Collapse
Affiliation(s)
- Yulong Liu
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Michelle E Kossack
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Matthew E McFaul
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Lana N Christensen
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Stefan Siebert
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Sydney R Wyatt
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Caramai N Kamei
- Mount Desert Island Biological LaboratoryBar HarborUnited States
| | - Samuel Horst
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Nayeli Arroyo
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Iain A Drummond
- Mount Desert Island Biological LaboratoryBar HarborUnited States
| | - Celina E Juliano
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| | - Bruce W Draper
- Department of Molecular and Cellular Biology, University of California, DavisDavisUnited States
| |
Collapse
|
7
|
Tran TN, Martinez J, Schimenti JC. A predicted deleterious allele of the essential meiosis gene MND1, present in ~ 3% of East Asians, does not disrupt reproduction in mice. Mol Hum Reprod 2020; 25:668-673. [PMID: 31393579 DOI: 10.1093/molehr/gaz048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/22/2019] [Accepted: 07/29/2019] [Indexed: 01/10/2023] Open
Abstract
Infertility is a major health problem affecting ~15% of couples worldwide. Except for cases involving readily detectable chromosome aberrations, confident identification of a causative genetic defect is problematic. Despite the advent of genome sequencing for diagnostic purposes, the preponderance of segregating genetic variants complicates identification of culprit genetic alleles or mutations. Many algorithms have been developed to predict the effects of 'variants of unknown significance', typically single nucleotide polymorphisms (SNPs), but these predictions are not sufficiently accurate for clinical action. As part of a project to identify population variants that impact fertility, we have been generating clustered regularly interspaced short palindromic repeats-Cas9 edited mouse models of suspect SNPs in genes that are known to be required for fertility in mice. Here, we present data on a non-synonymous (amino acid altering) SNP (rs140107488) in the meiosis gene Mnd1, which is predicted bioinformatically to be deleterious to protein function. We report that when modeled in mice, this allele (MND1K85M), which is present at an allele frequency of ~ 3% in East Asians, has no discernable effect upon fertility, fecundity or gametogenesis, although it may cause sex skewing of progeny from homozygous males. In sum, assuming the mouse model accurately reflects the impact of this variant in humans, rs140107488 appears to be a benign allele that can be eliminated or de-prioritized in clinical genomic analyses of infertility patients.
Collapse
Affiliation(s)
- Tina N Tran
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | | | - John C Schimenti
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
8
|
Nemani N, Dong Z, Daw CC, Madaris TR, Ramachandran K, Enslow BT, Rubannelsonkumar CS, Shanmughapriya S, Mallireddigari V, Maity S, SinghMalla P, Natarajanseenivasan K, Hooper R, Shannon CE, Tourtellotte WG, Singh BB, Reeves WB, Sharma K, Norton L, Srikantan S, Soboloff J, Madesh M. Mitochondrial pyruvate and fatty acid flux modulate MICU1-dependent control of MCU activity. Sci Signal 2020; 13:eaaz6206. [PMID: 32317369 PMCID: PMC7667998 DOI: 10.1126/scisignal.aaz6206] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The tricarboxylic acid (TCA) cycle converts the end products of glycolysis and fatty acid β-oxidation into the reducing equivalents NADH and FADH2 Although mitochondrial matrix uptake of Ca2+ enhances ATP production, it remains unclear whether deprivation of mitochondrial TCA substrates alters mitochondrial Ca2+ flux. We investigated the effect of TCA cycle substrates on MCU-mediated mitochondrial matrix uptake of Ca2+, mitochondrial bioenergetics, and autophagic flux. Inhibition of glycolysis, mitochondrial pyruvate transport, or mitochondrial fatty acid transport triggered expression of the MCU gatekeeper MICU1 but not the MCU core subunit. Knockdown of mitochondrial pyruvate carrier (MPC) isoforms or expression of the dominant negative mutant MPC1R97W resulted in increased MICU1 protein abundance and inhibition of MCU-mediated mitochondrial matrix uptake of Ca2+ We also found that genetic ablation of MPC1 in hepatocytes and mouse embryonic fibroblasts resulted in reduced resting matrix Ca2+, likely because of increased MICU1 expression, but resulted in changes in mitochondrial morphology. TCA cycle substrate-dependent MICU1 expression was mediated by the transcription factor early growth response 1 (EGR1). Blocking mitochondrial pyruvate or fatty acid flux was linked to increased autophagy marker abundance. These studies reveal a mechanism that controls the MCU-mediated Ca2+ flux machinery and that depends on TCA cycle substrate availability. This mechanism generates a metabolic homeostatic circuit that protects cells from bioenergetic crisis and mitochondrial Ca2+ overload during periods of nutrient stress.
Collapse
Affiliation(s)
- Neeharika Nemani
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Center for Translational Medicine, Lewis Katz School of Me.dicine at Temple University, Philadelphia, PA, 19140, USA
| | - Zhiwei Dong
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Center for Translational Medicine, Lewis Katz School of Me.dicine at Temple University, Philadelphia, PA, 19140, USA
| | - Cassidy C Daw
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Travis R Madaris
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Karthik Ramachandran
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Benjamin T Enslow
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Cherubina S Rubannelsonkumar
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Santhanam Shanmughapriya
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Center for Translational Medicine, Lewis Katz School of Me.dicine at Temple University, Philadelphia, PA, 19140, USA
- Heart and Vascular Institute, Department of Medicine and Department of Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, PA 17601, USA
| | - Varshini Mallireddigari
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Center for Translational Medicine, Lewis Katz School of Me.dicine at Temple University, Philadelphia, PA, 19140, USA
| | - Soumya Maity
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Pragya SinghMalla
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Kalimuthusamy Natarajanseenivasan
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Center for Translational Medicine, Lewis Katz School of Me.dicine at Temple University, Philadelphia, PA, 19140, USA
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Robert Hooper
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140, USA
| | - Christopher E Shannon
- Department of Medicine/Diabetes Division, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Warren G Tourtellotte
- Pathology & Laboratory Medicine, Neurology, Neurosurgery, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Brij B Singh
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - W Brian Reeves
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Kumar Sharma
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Luke Norton
- Department of Medicine/Diabetes Division, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Subramanya Srikantan
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jonathan Soboloff
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140, USA
| | - Muniswamy Madesh
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
- Center for Translational Medicine, Lewis Katz School of Me.dicine at Temple University, Philadelphia, PA, 19140, USA
- Department of Medicine/Nephrology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
9
|
Fang L, Zhou Y, Liu S, Jiang J, Bickhart DM, Null DJ, Li B, Schroeder SG, Rosen BD, Cole JB, Van Tassell CP, Ma L, Liu GE. Integrating Signals from Sperm Methylome Analysis and Genome-Wide Association Study for a Better Understanding of Male Fertility in Cattle. EPIGENOMES 2019; 3:epigenomes3020010. [PMID: 34968233 PMCID: PMC8594688 DOI: 10.3390/epigenomes3020010] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/03/2019] [Accepted: 05/11/2019] [Indexed: 01/18/2023] Open
Abstract
Decreased male fertility is a big concern in both human society and the livestock industry. Sperm DNA methylation is commonly believed to be associated with male fertility. However, due to the lack of accurate male fertility records (i.e., limited mating times), few studies have investigated the comprehensive impacts of sperm DNA methylation on male fertility in mammals. In this study, we generated 10 sperm DNA methylomes and performed a preliminary correlation analysis between signals from sperm DNA methylation and signals from large-scale (n = 27,214) genome-wide association studies (GWAS) of 35 complex traits (including 12 male fertility-related traits). We detected genomic regions, which experienced DNA methylation alterations in sperm and were associated with aging and extreme fertility phenotypes (e.g., sire-conception rate or SCR). In dynamic hypomethylated regions (HMRs) and partially methylated domains (PMDs), we found genes (e.g., HOX gene clusters and microRNAs) that were involved in the embryonic development. We demonstrated that genomic regions, which gained rather than lost methylations during aging, and in animals with low SCR were significantly and selectively enriched for GWAS signals of male fertility traits. Our study discovered 16 genes as the potential candidate markers for male fertility, including SAMD5 and PDE5A. Collectively, this initial effort supported a hypothesis that sperm DNA methylation may contribute to male fertility in cattle and revealed the usefulness of functional annotations in enhancing biological interpretation and genomic prediction for complex traits and diseases.
Collapse
Affiliation(s)
- Lingzhao Fang
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yang Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuli Liu
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jicai Jiang
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Derek M. Bickhart
- Dairy Forage Research Center, Agricultural Research Service, USDA, Madison, WI 53718, USA
| | - Daniel J. Null
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Bingjie Li
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Steven G. Schroeder
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Benjamin D. Rosen
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - John B. Cole
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Curtis P. Van Tassell
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Li Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
- Correspondence: (L.M.); (G.E.L.); Tel.: +1-301-405-1389 (L.M.); +1-301-504-9843 (G.E.L.); Fax: +1-301-504-8414 (G.E.L.)
| | - George E. Liu
- Animal Genomics and Improvement Laboratory, BARC, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
- Correspondence: (L.M.); (G.E.L.); Tel.: +1-301-405-1389 (L.M.); +1-301-504-9843 (G.E.L.); Fax: +1-301-504-8414 (G.E.L.)
| |
Collapse
|
10
|
Liao J, Ng SH, Luk AC, Suen HC, Qian Y, Lee AWT, Tu J, Fung JCL, Tang NLS, Feng B, Chan WY, Fouchet P, Hobbs RM, Lee TL. Revealing cellular and molecular transitions in neonatal germ cell differentiation using single cell RNA sequencing. Development 2019; 146:dev174953. [PMID: 30824552 DOI: 10.1242/dev.174953] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/17/2019] [Indexed: 12/22/2022]
Abstract
Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.
Collapse
Affiliation(s)
- Jinyue Liao
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Shuk Han Ng
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Alfred Chun Luk
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Hoi Ching Suen
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Yan Qian
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Annie Wing Tung Lee
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Jiajie Tu
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Jacqueline Chak Lam Fung
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Nelson Leung Sang Tang
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Bo Feng
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wai Yee Chan
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Joint CUHK-UoS (University of Southampton) Joint Laboratories for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- CUHK-BGI Innovation Institute of Trans-omics Hong Kong, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Pierre Fouchet
- CEA DRF IBFJ IRCM, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
- Université Paris Diderot, Sorbonne Paris Cité, INSERM, UMR 967, 92265 Fontenay-aux-Roses, France
- Université Paris Sud, INSERM, UMR 967, 92265 Fontenay-aux-Roses, France
| | - Robin M Hobbs
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Tin Lap Lee
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Joint CUHK-UoS (University of Southampton) Joint Laboratories for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- CUHK-BGI Innovation Institute of Trans-omics Hong Kong, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
11
|
Sinha D, Kalimutho M, Bowles J, Chan AL, Merriner DJ, Bain AL, Simmons JL, Freire R, Lopez JA, Hobbs RM, O'Bryan MK, Khanna KK. Cep55 overexpression causes male-specific sterility in mice by suppressing Foxo1 nuclear retention through sustained activation of PI3K/Akt signaling. FASEB J 2018; 32:4984-4999. [PMID: 29683733 DOI: 10.1096/fj.201701096rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spermatogenesis is a dynamic process involving self-renewal and differentiation of spermatogonial stem cells, meiosis, and ultimately, the differentiation of haploid spermatids into sperm. Centrosomal protein 55 kDa (CEP55) is necessary for somatic cell abscission during cytokinesis. It facilitates equal segregation of cytoplasmic contents between daughter cells by recruiting endosomal sorting complex required for transport machinery (ESCRT) at the midbody. In germ cells, CEP55, in partnership with testes expressed-14 (TEX14) protein, has also been shown to be an integral component of intercellular bridge before meiosis. Various in vitro studies have demonstrated a role for CEP55 in multiple cancers and other diseases. However, its oncogenic potential in vivo remains elusive. To investigate, we generated ubiquitously overexpressing Cep55 transgenic ( Cep55Tg/Tg) mice aiming to characterize its oncogenic role in cancer. Unexpectedly, we found that Cep55Tg/Tg male mice were sterile and had severe and progressive defects in spermatogenesis related to spermatogenic arrest and lack of spermatids in the testes. In this study, we characterized this male-specific phenotype and showed that excessively high levels of Cep55 results in hyperactivation of PI3K/protein kinase B (Akt) signaling in testis. In line with this finding, we observed increased phosphorylation of forkhead box protein O1 (FoxO1), and suppression of its nuclear retention, along with the relative enrichment of promyelocytic leukemia zinc finger (PLZF) -positive cells. Independently, we observed that Cep55 amplification favored upregulation of ret ( Ret) proto-oncogene and glial-derived neurotrophic factor family receptor α-1 ( Gfra1). Consistent with these data, we observed selective down-regulation of genes associated with germ cell differentiation in Cep55-overexpressing testes at postnatal day 10, including early growth response-4 ( Egr4) and spermatogenesis and oogenesis specific basic helix-loop-helix-1 ( Sohlh1). Thus, Cep55 amplification leads to a shift toward the initial maintenance of undifferentiated spermatogonia and ultimately results in progressive germ cell loss. Collectively, our findings demonstrate that Cep55 overexpression causes change in germ cell proportions and manifests as a Sertoli cell only tubule phenotype, similar to that seen in many azoospermic men.-Sinha, D., Kalimutho, M., Bowles, J., Chan, A.-L., Merriner, D. J., Bain, A. L., Simmons, J. L., Freire, R., Lopez, J. A., Hobbs, R. M., O'Bryan, M. K., Khanna, K. K. Cep55 overexpression causes male-specific sterility in mice by suppressing Foxo1 nuclear retention through sustained activation of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Debottam Sinha
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Murugan Kalimutho
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Ai-Leen Chan
- Germline Stem Cell Laboratory, Australian Regenerative Medicine Institute and Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - D Jo Merriner
- Male Infertility and Germ Cell Biology Laboratory, the School of Biological Sciences, Monash University, Clayton, Victoria, Australia; and
| | - Amanda L Bain
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jacinta L Simmons
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Tenerife, Spain
| | - J Alejandro Lopez
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Robin M Hobbs
- Germline Stem Cell Laboratory, Australian Regenerative Medicine Institute and Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Moira K O'Bryan
- Male Infertility and Germ Cell Biology Laboratory, the School of Biological Sciences, Monash University, Clayton, Victoria, Australia; and
| | - Kum Kum Khanna
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Queensland, Australia
| |
Collapse
|
12
|
Kim HR, Kim YS, Yoon JA, Yang SC, Park M, Seol DW, Lyu SW, Jun JH, Lim HJ, Lee DR, Song H. Estrogen induces EGR1 to fine-tune its actions on uterine epithelium by controlling PR signaling for successful embryo implantation. FASEB J 2018; 32:1184-1195. [PMID: 29092905 DOI: 10.1096/fj.201700854rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The harmonized actions of ovarian E2 and progesterone (P4) regulate the proliferation and differentiation of uterine cells in a spatiotemporal manner. Imbalances between these hormones often lead to infertility and gynecologic diseases. Whereas numerous factors that are involved in P4 signaling have been identified, few local factors that mediate E2 actions in the uterus have been revealed. Here, we demonstrate that estrogen induces the transcription factor, early growth response 1 ( Egr1), to fine-tune its actions in uterine epithelial cells (ECs) that are responsible for uterine receptivity for embryo implantation. In the presence of exogenous gonadotrophins, ovulation, fertilization, and embryonic development normally occur in Egr1-/- mice, but these animals experience the complete failure of embryo implantation with reduced artificial decidualization. Although serum levels of E2 and P4 were comparable between Egr1+/+ and Egr1-/- mice on d 4 of pregnancy, aberrantly reduced levels of progesterone receptor in Egr1-/- uterine ECs caused enhanced E2 activity and impaired P4 response. Ultrastructural analyses revealed that Egr1-/- ECs are not fully able to provide proper uterine receptivity. Uterine mRNA landscapes in Egr1-/- mice revealed that EGR1 controls the expression of a subset of E2-regulated genes. In addition, P4 signaling was unable to modulate estrogen actions, including those that are involved in cell-cycle progression, in ECs that were deficient in EGR1. Furthermore, primary coculture of Egr1-/- ECs with Egr1+/+ stromal cells, and vice versa, supported the notion that Egr1 is required to modulate E2 actions on ECs to prepare the uterine environment for embryo implantation. In contrast to its role in ECs, loss of Egr1 in stroma significantly reduced stromal cell proliferation. Collectively, our results demonstrate that E2 induces EGR1 to streamline its actions for the preparation of uterine receptivity for embryo implantation in mice.-Kim, H.-R., Kim, Y. S., Yoon, J. A., Yang, S. C., Park, M., Seol, D.-W., Lyu, S. W., Jun, J. H., Lim, H. J., Lee, D. R., Song, H. Estrogen induces EGR1 to fine-tune its actions on uterine epithelium by controlling PR signaling for successful embryo implantation.
Collapse
Affiliation(s)
- Hye-Ryun Kim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Jung Ah Yoon
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Seung Chel Yang
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Dong-Won Seol
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Sang Woo Lyu
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Jin Hyun Jun
- Department of Biomedical Laboratory Science, Graduate School of Health Science, Eulji University, Seongnam, Korea
| | | | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Korea
| |
Collapse
|
13
|
Han P, Guerrero-Netro H, Estienne A, Cao B, Price CA. Regulation and action of early growth response 1 in bovine granulosa cells. Reproduction 2017; 154:547-557. [PMID: 28733346 DOI: 10.1530/rep-17-0243] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 06/26/2017] [Accepted: 07/21/2017] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factors (FGF) modify cell proliferation and differentiation through receptor tyrosine kinases, which stimulate the expression of transcription factors including members of the early growth response (EGR) family. In ovarian granulosa cells, most FGFs activate typical response genes, although the role of EGR proteins has not been described. In the present study, we determined the regulation of EGR mRNA by FGFs and explored the role of EGR1 in the regulation of FGF-response genes. Addition of FGF1, FGF2, FGF4 or FGF8b increased EGR1 and EGR3 mRNA levels, whereas FGF18 increased only EGR1 mRNA abundance. No mRNA encoding EGR2 or EGR4 was detected. Overexpression of EGR1 increased EGR3 mRNA levels as well as the FGF-response genes SPRY2, NR4A1 and FOSL1 and also increased the phosphorylation of MAPK3/1. Knockdown of EGR3 did not alter the ability of FGF8b to stimulate SPRY2 mRNA levels. These data demonstrate the regulation of EGR1 and EGR3 mRNA abundance by FGFs in granulosa cells and suggest that EGR1 is likely an upstream component of FGF signaling in granulosa cells.
Collapse
Affiliation(s)
- Peng Han
- College of Animal Science and TechnologyNorthwest A&F University, Yangling, Shaanxi, China.,Centre de Recherche en Reproduction et Fertilité (CRRF)Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Hilda Guerrero-Netro
- Centre de Recherche en Reproduction et Fertilité (CRRF)Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Anthony Estienne
- Centre de Recherche en Reproduction et Fertilité (CRRF)Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| | - Binyun Cao
- College of Animal Science and TechnologyNorthwest A&F University, Yangling, Shaanxi, China
| | - Christopher A Price
- Centre de Recherche en Reproduction et Fertilité (CRRF)Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Quebec, Canada
| |
Collapse
|
14
|
Santino P, Martignani E, Miretti S, Baratta M, Accornero P. Mechanisms of modulation of the Egr gene family in mammary epithelial cells of different species. Gen Comp Endocrinol 2017; 247:87-96. [PMID: 28118985 DOI: 10.1016/j.ygcen.2017.01.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/10/2017] [Accepted: 01/20/2017] [Indexed: 10/20/2022]
Abstract
In the adult female, within the estrous cycle, the mammary gland undergoes multiple rounds of growth, with increased cellular proliferation, and involution, with increased apoptosis. The increase in proliferation is elicited by endocrine (Estrogen, Progesterone), as well as locally produced (epidermal growth factor, insulin-like growth factor, etc) growth factors. Among the genes that are modulated during cellular proliferation, immediate early genes play a fundamental role, being rapidly upregulated and then downregulated within the G0/G1 phase of the cell cycle, allowing the progression to the subsequent phases. Egrs (1-4) are immediate early genes that encode for transcription factors that promote, within different cell types and depending on the strength and duration of the stimuli, several different responses like mitogenesis, differentiation, apoptosis or even anti-apoptosis. In this work we have studied the mechanisms of modulation of the Egr family, in mammary epithelial cells of different origin (bovine, canine, feline, murine). Following stimulation with growth medium, Egr mRNA expression showed a strong upregulation reaching a peak at 45-60min, that rapidly declined. Among several cytokines, particularly important for mammary morphogenesis, that we have tested (EGF, IGF-I, insulin, estrogen, progesterone), only EGF upregulated Egrs to levels close to those elicited by growth medium. In order to understand how the Egr transcription factors were regulated, we have inhibited Erk 1/2 and PI3K, molecules that drive two major intracellular signaling pathways. Inhibition of the Erk 1/2 pathway totally abolished Egr upregulation mediated by growth medium or EGF. On the other hand, the PI3K-Akt pathway played a minor role on Egr levels, with a strong inhibitory effect on cat GH2 cells only, that could be ascribed to reduced Erk phosphorylation following PI3K inhibition. Finally we showed that addition of growth medium also upregulated that the mammary luminal marker cytokeratin 18, but only in the murine NMuMG cell line. This is the first manuscript describing how the Egr transcription factors are expressed in mammary epithelial cells of domestic animals and which growth factors and signaling pathways modulate their expression.
Collapse
Affiliation(s)
- P Santino
- Dipartimento di Scienze Veterinarie, Largo Braccini 2, 10095 Grugliasco (TO), Italy.
| | - E Martignani
- Dipartimento di Scienze Veterinarie, Largo Braccini 2, 10095 Grugliasco (TO), Italy.
| | - S Miretti
- Dipartimento di Scienze Veterinarie, Largo Braccini 2, 10095 Grugliasco (TO), Italy.
| | - M Baratta
- Dipartimento di Scienze Veterinarie, Largo Braccini 2, 10095 Grugliasco (TO), Italy.
| | - P Accornero
- Dipartimento di Scienze Veterinarie, Largo Braccini 2, 10095 Grugliasco (TO), Italy.
| |
Collapse
|
15
|
Sung SR, Song SH, Kang KM, Park JE, Nam YJ, Shin YJ, Cha DH, Seo JT, Yoon TK, Shim SH. Sequence variations of the EGR4 gene in Korean men with spermatogenesis impairment. BMC MEDICAL GENETICS 2017; 18:47. [PMID: 28464846 PMCID: PMC5414287 DOI: 10.1186/s12881-017-0408-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 04/19/2017] [Indexed: 11/17/2022]
Abstract
Background Egr4 is expressed in primary and secondary spermatocytes in adult mouse testes and has a crucial role in regulating germ cell maturation. The functional loss of Egr4 blocks spermatogenesis, significantly reducing the number of spermatozoa that are produced. In this study, we examined whether EGR4 variants are present in Korean men with impaired spermatogenesis. Methods A total 170 Korean men with impaired spermatogenesis and 272 normal controls were screened. The coding regions including exon-intron boundaries of EGR4 were sequenced by PCR-direct sequencing method. Results We identified eight sequence variations in the coding region and 3′-UTR regions of the EGR4 gene. Four were nonsynonymous variants (rs771189047, rs561568849, rs763487015, and rs546250227), three were synonymous variants (rs115948271, rs528939702, and rs7558708), and one variant (rs2229294) was localized in the 3′-UTR. Three nonsynonymous variants [c.65_66InsG (p. Cys23Leufs*37), c.236C > T (p. Pro79Leu), c.1294G > T (p. Val432Leu)] and one synonymous variant [c.1230G > A (p. Thr410)] were not detected in controls. To evaluate the pathogenic effects of nonsynonymous variants, we used seven prediction methods. The c.214C > A (p. Arg72Ser) and c.236C > T (p. Pro79Leu) variants were predicted as “damaging” by SIFT and SNAP2. The c.65_66insG (p. Cys23Leufs*37) variants were predicted as “disease causing” by Mutation Taster, SNPs &GO and SNAP2. The c.867C > G (p. Leu289) variants were predicted as “disease causing” only by Mutation Taster. Conclusion To date, this study is the first to screen the EGR4 gene in relation to male infertility. However, our findings did not clearly explain how nonsynonymous EGR4 variations affect spermatogenesis. Therefore, further studies are required to validate the functional impact of EGR4 variations on spermatogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12881-017-0408-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Se Ra Sung
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, Seoul, South Korea
| | - Seung Hun Song
- Department of Urology, CHA Gangnam Medical Center, Seoul, South Korea
| | - Kyung Min Kang
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, Seoul, South Korea
| | - Ji Eun Park
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, Seoul, South Korea
| | - Yeo Jung Nam
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, South Korea
| | - Yun-Jeong Shin
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, Seoul, South Korea
| | - Dong Hyun Cha
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center, Seoul, South Korea
| | - Ju Tae Seo
- Department of Urology, Cheil General Hospital, Seoul, South Korea
| | - Tae Ki Yoon
- Department of Obstetrics and Gynecology, CHA Gangnam Medical Center, Seoul, South Korea
| | - Sung Han Shim
- Genetics Laboratory, Fertility Center of CHA Gangnam Medical Center, Seoul, South Korea. .,Department of Biomedical Science, College of Life Science, CHA University, Seoul, South Korea.
| |
Collapse
|
16
|
Zhang X, Huang L, Wu T, Feng Y, Ding Y, Ye P, Yin Z. Transcriptomic Analysis of Ovaries from Pigs with High And Low Litter Size. PLoS One 2015; 10:e0139514. [PMID: 26426260 PMCID: PMC4591126 DOI: 10.1371/journal.pone.0139514] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/13/2015] [Indexed: 01/03/2023] Open
Abstract
Litter size is one of the most important economic traits for pig production as it is directly related to the production efficiency. Litter size is affected by interactions between multiple genes and the environment. While recent studies have identified some genes associated with prolificacy in pigs, transcriptomic studies of specific genes affecting litter size in porcine ovaries are rare. In order to identify candidate genes associated with litter size in swine, we assessed gene expression differences between the ovaries of Yorkshire pigs with extremely high and low litter sizes using the RNA-Seq method. A total of 1 243 differentially expressed genes were identified: 897 genes were upregulated and 346 genes were downregulated in high litter size ovary samples compared with low litter size ovary samples. A large number of these genes related to steroid hormone regulation in animal ovaries, including 59 Gene Ontology terms and 27 Kyoto Encyclopedia of Genes and Genomes pathways involved in steroid biosynthesis and ovarian steroidogenesis. From these differentially expressed genes, we identified a total of 11 genes using a bioinformatics screen that may be associated with high litter size in Yorkshire pigs. These results provide a list of new candidate genes for porcine litter size and prolificacy to be further investigated.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
| | - Long Huang
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
| | - Tao Wu
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
| | - Yifang Feng
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
| | - Yueyun Ding
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
| | - Pengfei Ye
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
| | - Zongjun Yin
- Key Laboratory of Local Animal Genetic Resources Conservation and Bio-breeding of Anhui province, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui Province, People’s Republic of China
- * E-mail:
| |
Collapse
|
17
|
A novel function for Egr4 in posterior hindbrain development. Sci Rep 2015; 5:7750. [PMID: 25583070 PMCID: PMC4291570 DOI: 10.1038/srep07750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/09/2014] [Indexed: 02/08/2023] Open
Abstract
Segmentation of the vertebrate hindbrain is an evolutionarily conserved process. Here, we identify the transcription factor early growth response 4 (egr4) as a novel regulator of posterior hindbrain development in Xenopus. egr4 is specifically and transiently expressed in rhombomeres 5 and 6 (r5/r6), and Egr4 knockdown causes a loss of mafb/kreisler and krox20/egr2 expression in r5/r6 and r5, respectively. This phenotype can be fully rescued by injection of frog or mouse Egr4 mRNA. Moreover Egr4-depleted embryos exhibit a specific loss of the neural crest stream adjacent to r5, and have inner ear defects. While the homeodomain protein vHnf1/Hnf1b directly activates Mafb and Krox20 expression in the mouse hindbrain to specify r5, we show that in Xenopus this process is indirect through the activation of Egr4. We provide evidence that rearrangements in the regulatory sequences around egr4 and mafb genes may account for this difference.
Collapse
|
18
|
Wang J, Zhao Y, Gu K, Yu P, Zhang B, Wang W, Yang J, Xu Y. The novel porcine gene early growth response 4 (Egr4) is differentially expressed in the ovaries of Erhualian and Pietrain pigs. Reprod Fertil Dev 2014; 26:587-98. [PMID: 23719176 DOI: 10.1071/rd12380] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/28/2013] [Indexed: 01/09/2023] Open
Abstract
The early growth response 4 (Egr4) gene plays a critical role in human and mouse fertility. In the present study, Affymetrix microarray gene chips were used to evaluate differential gene expression in the ovaries between Erhualian and Pietrain pigs. In all, 487 and 573 transcripts were identified with significantly higher and lower expression, respectively, in Erhualian compared with Pietrain sows. The Egr4 gene, one of the differentially expressed genes, was cloned and its genomic structure was analysed. Egr4 expression is increased 120-fold in ovaries from Erhualian sows. The full-length cDNA of porcine Egr4 was obtained by in silico cloning and 5' rapid amplification of cDNA ends. The gene consists of two exons and its predicted protein contains a Cys2His2 zinc finger structure. The porcine transcript is alternatively spliced by exon sequence deletion, producing two different mRNAs differing at the 5' end of Exon 2. Egr4 transcripts were detected in the central nervous system, including the cerebrum, cerebellum, hypothalamus and pituitary gland, and were highly expressed in the ovary. The Egr4 gene was evaluated as a candidate gene for porcine reproductivity. To investigate the role of Egr4 in the ovary, Egr4 was knocked down using short interference (si) RNA in porcine granulosa cells. Knockdown of Egr4 using siRNA effectively inhibited Egr4 mRNA and protein expression and knockdown significantly affected the expression of Bax, P450arom, P450scc, Egr1, Egr2, and Egr3. In conclusion, these observations establish an important role for Egr4 in the porcine ovary.
Collapse
Affiliation(s)
- Jingjing Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yongyan Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kecui Gu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Baole Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Juanjuan Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yinxue Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
19
|
Matsuo T, Dat LT, Komatsu M, Yoshimaru T, Daizumoto K, Sone S, Nishioka Y, Katagiri T. Early growth response 4 is involved in cell proliferation of small cell lung cancer through transcriptional activation of its downstream genes. PLoS One 2014; 9:e113606. [PMID: 25411851 PMCID: PMC4239076 DOI: 10.1371/journal.pone.0113606] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022] Open
Abstract
Small cell lung cancer (SCLC) is aggressive, with rapid growth and frequent bone metastasis; however, its detailed molecular mechanism remains poorly understood. Here, we report the critical role of early growth factor 4 (EGR4), a DNA-binding, zinc-finger transcription factor, in cell proliferation of SCLC. EGR4 overexpression in HEK293T cells conferred significant upregulation of specific splice variants of the parathyroid hormone-related protein (PTHrP) gene, resulting in enhancement of the secretion of PTHrP protein, a known mediator of osteolytic bone metastasis. More importantly, depletion of EGR4 expression by siRNA significantly suppressed growth of the SCLC cell lines, SBC-5, SBC-3 and NCI-H1048. On the other hand, introduction of EGR4 into NIH3T3 cells significantly enhanced cell growth. We identified four EGR4 target genes, SAMD5, RAB15, SYNPO and DLX5, which were the most significantly downregulated genes upon depletion of EGR4 expression in all of the SCLC cells examined, and demonstrated the direct recruitment of EGR4 to their promoters by ChIP and luciferase reporter analysis. Notably, knockdown of the expression of these genes by siRNA remarkably suppressed the growth of all the SCLC cells. Taken together, our findings suggest that EGR4 likely regulates the bone metastasis and proliferation of SCLC cells via transcriptional regulation of several target genes, and may therefore be a promising target for the development of anticancer drugs for SCLC patients.
Collapse
Affiliation(s)
- Taisuke Matsuo
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| | - Le Tan Dat
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
- Department of Medical Oncology, Institute of Health Biosciences, The University of Tokushima, Tokushima, Japan
| | - Masato Komatsu
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| | - Tetsuro Yoshimaru
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| | - Kei Daizumoto
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
| | - Saburo Sone
- Department of Medical Oncology, Institute of Health Biosciences, The University of Tokushima, Tokushima, Japan
| | - Yasuhiko Nishioka
- Department of Medical Oncology, Institute of Health Biosciences, The University of Tokushima, Tokushima, Japan
| | - Toyomasa Katagiri
- Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Tokushima, Japan
- * E-mail:
| |
Collapse
|
20
|
Liang XH, Deng WB, Li M, Zhao ZA, Wang TS, Feng XH, Cao YJ, Duan EK, Yang ZM. Egr1 protein acts downstream of estrogen-leukemia inhibitory factor (LIF)-STAT3 pathway and plays a role during implantation through targeting Wnt4. J Biol Chem 2014; 289:23534-45. [PMID: 25012664 DOI: 10.1074/jbc.m114.588897] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Embryo implantation is a highly synchronized process between an activated blastocyst and a receptive uterus. Successful implantation relies on the dynamic interplay of estrogen and progesterone, but the key mediators underlying embryo implantation are not fully understood. Here we show that transcription factor early growth response 1 (Egr1) is regulated by estrogen as a downstream target through leukemia inhibitory factor (LIF) signal transducer and activator of transcription 3 (STAT3) pathway in mouse uterus. Egr1 is localized in the subluminal stromal cells surrounding the implanting embryo on day 5 of pregnancy. Estrogen rapidly, markedly, and transiently enhances Egr1 expression in uterine stromal cells, which fails in estrogen receptor α knock-out mouse uteri. STAT3 is phosphorylated by LIF and subsequently recruited on Egr1 promoter to induce its expression. Our results of Egr1 expression under induced decidualization in vivo and in vitro show that Egr1 is rapidly induced after deciduogenic stimulus. Egr1 knockdown can inhibit in vitro decidualization of cultured uterine stromal cells. Chromatin immunoprecipitation data show that Egr1 is recruited to the promoter of wingless-related murine mammary tumor virus integration site 4 (Wnt4). Collectively, our study presents for the first time that estrogen regulates Egr1 expression through LIF-STAT3 signaling pathway in mouse uterus, and Egr1 functions as a critical mediator of stromal cell decidualization by regulating Wnt4.
Collapse
Affiliation(s)
- Xiao-Huan Liang
- From the College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wen-Bo Deng
- From the College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ming Li
- College of Life Science, Xiamen University, Xiamen 361005, China
| | - Zhen-Ao Zhao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing 100101, China, and
| | - Tong-Song Wang
- Department of Biology, Shantou University, Shantou 515063, China
| | - Xu-Hui Feng
- College of Life Science, Xiamen University, Xiamen 361005, China
| | - Yu-Jing Cao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing 100101, China, and
| | - En-Kui Duan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing 100101, China, and
| | - Zeng-Ming Yang
- From the College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China,
| |
Collapse
|
21
|
Shin H, Kwon S, Song H, Lim HJ. The transcription factor Egr3 is a putative component of the microtubule organizing center in mouse oocytes. PLoS One 2014; 9:e94708. [PMID: 24722338 PMCID: PMC3983223 DOI: 10.1371/journal.pone.0094708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/17/2014] [Indexed: 11/19/2022] Open
Abstract
The early growth response (Egr) family of zinc finger transcription factors consists of 4 members. During an investigation of Egr factor localization in mouse ovaries, we noted that Egr3 exhibits a subcellular localization that overlaps with the meiotic spindle in oocytes. Using Egr3-specific antibodies, we establish that Egr3 co-localizes with the spindle and cytosolic microtubule organizing centers (MTOCs) in oocytes during meiotic maturation. Notably, the Egr3 protein appears to accumulate around γ-tubulin in MTOCs. Nocodazole treatment, which induces microtubule depolymerization, resulted in the disruption of spindle formation and Egr3 localization, suggesting that Egr3 localization is dependent on the correct configuration of the spindle. Shortly after warming of vitrified oocytes, growing arrays of microtubules were observed near large clusters of Egr3. An in vitro microtubule interaction assay showed that Egr3 does not directly interact with polymerized microtubules. Egr3 localization on the spindle was sustained in early preimplantation mouse embryos, but this pattern did not persist until the blastocyst stage. Collectively, our result shows for the first time that the Egr3 a transcription factor may play a novel non-transcriptional function during microtubule organization in mouse oocytes.
Collapse
Affiliation(s)
- Hyejin Shin
- Department of Biomedical Science & Technology, Institute of Biomedical Science & Technology, Konkuk University, Seoul, Korea
| | - Sojung Kwon
- Department of Biomedical Science & Technology, Institute of Biomedical Science & Technology, Konkuk University, Seoul, Korea
| | - Haengseok Song
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea
| | - Hyunjung Jade Lim
- Department of Biomedical Science & Technology, Institute of Biomedical Science & Technology, Konkuk University, Seoul, Korea
- * E-mail:
| |
Collapse
|
22
|
Ng JH, Kumar V, Muratani M, Kraus P, Yeo JC, Yaw LP, Xue K, Lufkin T, Prabhakar S, Ng HH. In vivo epigenomic profiling of germ cells reveals germ cell molecular signatures. Dev Cell 2013; 24:324-33. [PMID: 23352811 DOI: 10.1016/j.devcel.2012.12.011] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 10/17/2012] [Accepted: 12/19/2012] [Indexed: 12/01/2022]
Abstract
The limited number of in vivo germ cells poses an impediment to genome-wide studies. Here, we applied a small-scale chromatin immunoprecipitation sequencing (ChIP-seq) method on purified mouse fetal germ cells to generate genome-wide maps of four histone modifications (H3K4me3, H3K27me3, H3K27ac, and H2BK20ac). Comparison of active chromatin state between somatic, embryonic stem, and germ cells revealed promoters and enhancers needed for stem cell maintenance and germ cell development. We found the nuclear receptor Nr5a2 motif to be enriched at a subset of germ cell cis-regulatory regions, and our results implicate Nr5a2 in germ cell biology. Interestingly, in germ cells, the H3K27me3 histone modification occurs more frequently at regions that are enriched for retrotransposons and MHC genes, indicating that these loci are specifically silenced in germ cells. Together, our study provides genome-wide histone modification maps of in vivo germ cells and reveals the molecular chromatin signatures of germ cells.
Collapse
Affiliation(s)
- Jia-Hui Ng
- Gene Regulation Laboratory, Genome Institute of Singapore, Singapore 138672
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chalmel F, Lardenois A, Evrard B, Mathieu R, Feig C, Demougin P, Gattiker A, Schulze W, Jégou B, Kirchhoff C, Primig M. Global human tissue profiling and protein network analysis reveals distinct levels of transcriptional germline-specificity and identifies target genes for male infertility. Hum Reprod 2012; 27:3233-48. [PMID: 22926843 DOI: 10.1093/humrep/des301] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Mammalian spermatogenesis is a process that involves a complex expression program in both somatic and germ cells present in the male gonad. A number of studies have attempted to define the transcriptome of male meiosis and gametogenesis in rodents and primates. Few human transcripts, however, have been associated with testicular somatic cells and germ cells at different post-natal developmental stages and little is known about their level of germline-specificity compared with non-testicular tissues. METHODS We quantified human transcripts using GeneChips and a total of 47 biopsies from prepubertal children diagnosed with undescended testis, infertile adult patients whose spermatogenesis is arrested at consecutive stages and fertile control individuals. These results were integrated with data from enriched normal germ cells, non-testicular expression data, phenotype information, predicted regulatory DNA-binding motifs and interactome data. RESULTS Among 3580 genes for which we found differential transcript concentrations in somatic and germ cells present in human testis, 933 were undetectable in 45 embryonic and adult non-testicular tissues, including many that were corroborated at protein level by published gene annotation data and histological high-throughput protein immunodetection assays. Using motif enrichment analyses, we identified regulatory promoter elements likely involved in germline development. Finally, we constructed a regulatory disease network for human fertility by integrating expression signals, interactome information, phenotypes and functional annotation data. CONCLUSIONS Our results provide broad insight into the post-natal human testicular transcriptome at the level of cell populations and in a global somatic tissular context. Furthermore, they yield clues for genetic causes of male infertility and will facilitate the identification of novel cancer/testis genes as targets for cancer immunotherapies.
Collapse
Affiliation(s)
- Frédéric Chalmel
- Inserm Unit 1085-IRSET, Université de Rennes 1, EHESP School of Public Health, F-35042 Rennes, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Burns WR, Sabanegh E, Dada R, Rein B, Agarwal A. Is male infertility a forerunner to cancer? Int Braz J Urol 2011; 36:527-36. [PMID: 21044369 DOI: 10.1590/s1677-55382010000500002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2010] [Indexed: 11/22/2022] Open
Abstract
PURPOSE The frequency of testicular cancer and male infertility has been increasing in the past several decades. This article examines the relationship between male infertility and testicular cancer, concentrating particularly on causal links. RESULTS Both of these disorders are associated with testicular dysgenesis syndrome and have also been traced to mutations in genes involving DNA repair and tumor suppression, as well as environmental exposure. CONCLUSION The identification and examination of these common points of origin supports the integration of testicular cancer screenings into the routine evaluation of infertile men.
Collapse
Affiliation(s)
- Whitney R Burns
- Center for Reproductive Medicine, Glickman Urological, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
25
|
Goertz MJ, Wu Z, Gallardo TD, Hamra FK, Castrillon DH. Foxo1 is required in mouse spermatogonial stem cells for their maintenance and the initiation of spermatogenesis. J Clin Invest 2011; 121:3456-66. [PMID: 21865646 DOI: 10.1172/jci57984] [Citation(s) in RCA: 206] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 07/06/2011] [Indexed: 12/16/2022] Open
Abstract
Spermatogonial stem cells (SSCs) capable of self-renewal and differentiation are the foundation for spermatogenesis. Although several factors important for these processes have been identified, the fundamental mechanisms regulating SSC self-renewal and differentiation remain unknown. Here, we investigated a role for the Foxo transcription factors in mouse spermatogenesis and found that Foxo1 specifically marks mouse gonocytes and a subset of spermatogonia with stem cell potential. Genetic analyses showed that Foxo1 was required for both SSC homeostasis and the initiation of spermatogenesis. Combined deficiency of Foxo1, Foxo3, and Foxo4 resulted in a severe impairment of SSC self-renewal and a complete block of differentiation, indicating that Foxo3 and Foxo4, although dispensable for male fertility, contribute to SSC function. By conditional inactivation of 3-phosphoinositide-dependent protein kinase 1 (Pdk1) and phosphatase and tensin homolog (Pten) in the male germ line, we found that PI3K signaling regulates Foxo1 stability and subcellular localization, revealing that the Foxos are pivotal effectors of PI3K-Akt signaling in SSCs. We also identified a network of Foxo gene targets--most notably Ret--that rationalized the maintenance of SSCs by the Foxos. These studies demonstrate that Foxo1 expression in the spermatogenic lineage is intimately associated with the stem cell state and revealed what we believe to be novel Foxo-dependent mechanisms underlying SSC self-renewal and differentiation, with implications for common diseases, including male infertility and testicular cancer, due to abnormalities in SSC function.
Collapse
Affiliation(s)
- Meredith J Goertz
- Department of Pathology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, Texas 75390-9072, USA
| | | | | | | | | |
Collapse
|
26
|
Hogarth CA, Mitchell D, Small C, Griswold M. EGR4 displays both a cell- and intracellular-specific localization pattern in the developing murine testis. Dev Dyn 2011; 239:3106-14. [PMID: 20925118 DOI: 10.1002/dvdy.22442] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Spermatogenesis is an intricately regulated process of cellular differentiation transforming spermatogonial stem cells to spermatozoa. Elimination of the transcription factor EGR4 generates subfertile male mice yet the expression and function of EGR4 in the mammalian testis has yet to be fully investigated. We performed in situ hybridization and immunofluorescence to identify Egr4 transcript and protein localization in the developing murine testis. EGR4 was detected in both germ and somatic cells in the neonatal testis but was specific to germ cells inside the seminiferous epithelium from juvenile development onward. EGR4 also displayed distinct intracellular localization patterns within specific cell populations of the testis. In addition, Egr4-deficient testis tubules regress from relatively normal to Sertoli cell and undifferentiated spermatogonia only over time. Taken together, these data suggest that Egr4 may regulate spermatogenesis at multiple steps, with roles in the dividing Sertoli cells, peritubular myoid cells, and the meiotic and elongating haploid germ cell populations.
Collapse
Affiliation(s)
- Cathryn A Hogarth
- School of Molecular Biosciences and Centre for Reproductive Biology, Washington State University, Pullman, Washington 99164, USA
| | | | | | | |
Collapse
|
27
|
Clary LM, Okkema PG. The EGR family gene egrh-1 functions non-autonomously in the control of oocyte meiotic maturation and ovulation in C. elegans. Development 2010; 137:3129-37. [PMID: 20736289 PMCID: PMC2926961 DOI: 10.1242/dev.041616] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2010] [Indexed: 01/31/2023]
Abstract
Oocyte production, maturation and ovulation must be coordinated with sperm availability for successful fertilization. In C. elegans this coordination involves signals from the sperm to the oocyte and somatic gonad, which stimulate maturation and ovulation. We have found that the C. elegans early growth response factor family member EGRH-1 inhibits oocyte maturation and ovulation until sperm are available. In the absence of sperm, egrh-1 mutants exhibit derepressed oocyte maturation marked by MAPK activation and ovulation. egrh-1 mutants exhibit ectopic oocyte differentiation in the distal gonadal arm and accumulate abnormal and degraded oocytes proximally. These defects result in reduced brood size and partially penetrant embryonic lethality. We have found that endogenous EGRH-1 protein and an egrh-1::gfp reporter gene are expressed in the sheath and distal tip cells of the somatic gonad, the gut and other non-gonadal tissues, as well as in sperm, but expression is not observed in oocytes. Results of tissue-specific egrh-1(RNAi) experiments and genetic mosaic analyses revealed that EGRH-1 function is necessary in the soma and, surprisingly, this function is required in both the gut and the somatic gonad. Based on transformation rescue experiments we hypothesize that EGRH-1 in the somatic gonad inhibits oocyte maturation and ovulation.
Collapse
Affiliation(s)
- Lynn M. Clary
- Laboratory for Molecular Biology, Department of Biological Sciences, University of Illinois at Chicago, 900 South Ashland Avenue (MC567), Chicago, IL 60607, USA
| | - Peter G. Okkema
- Laboratory for Molecular Biology, Department of Biological Sciences, University of Illinois at Chicago, 900 South Ashland Avenue (MC567), Chicago, IL 60607, USA
| |
Collapse
|
28
|
Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 4: intercellular bridges, mitochondria, nuclear envelope, apoptosis, ubiquitination, membrane/voltage-gated channels, methylation/acetylation, and transcription factors. Microsc Res Tech 2010; 73:364-408. [PMID: 19941288 DOI: 10.1002/jemt.20785] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
As germ cells divide and differentiate from spermatogonia to spermatozoa, they share a number of structural and functional features that are common to all generations of germ cells and these features are discussed herein. Germ cells are linked to one another by large intercellular bridges which serve to move molecules and even large organelles from the cytoplasm of one cell to another. Mitochondria take on different shapes and features and topographical arrangements to accommodate their specific needs during spermatogenesis. The nuclear envelope and pore complex also undergo extensive modifications concomitant with the development of germ cell generations. Apoptosis is an event that is normally triggered by germ cells and involves many proteins. It occurs to limit the germ cell pool and acts as a quality control mechanism. The ubiquitin pathway comprises enzymes that ubiquitinate as well as deubiquitinate target proteins and this pathway is present and functional in germ cells. Germ cells express many proteins involved in water balance and pH control as well as voltage-gated ion channel movement. In the nucleus, proteins undergo epigenetic modifications which include methylation, acetylation, and phosphorylation, with each of these modifications signaling changes in chromatin structure. Germ cells contain specialized transcription complexes that coordinate the differentiation program of spermatogenesis, and there are many male germ cell-specific differences in the components of this machinery. All of the above features of germ cells will be discussed along with the specific proteins/genes and abnormalities to fertility related to each topic.
Collapse
Affiliation(s)
- Louis Hermo
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, 3640 University Street, Montreal, QC Canada H3A 2B2.
| | | | | | | |
Collapse
|
29
|
Liu BC, Zhang J, Wang L, Li XW, Wang Y, Ji J, Yang FP, Wan CL, Gao LH, Xu YF, Feng GY, He L, Zhao XZ, He G. No association between EGR gene family polymorphisms and schizophrenia in the Chinese population. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:506-9. [PMID: 20144677 DOI: 10.1016/j.pnpbp.2010.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 12/13/2022]
Abstract
Early growth response (EGR) genes are thought to have a role in the pathogenesis of schizophrenia because of their conserved DNA binding domain and biologically activity in neuronal plasticity. This zinc-finger motif could influence gene post-translational modification and expression. The multigenetic association model, using markers in genes of similar or antagonistic biological effects within a signal pathway or gene family, might be more appropriate to this aspect of the schizophrenia hypothesis than the single gene strategy. In this study we investigated the role of EGR1, EGR2, EGR3 and EGR4 within the EGR family. Taqman technology was used to examine 12 single nucleotide polymorphisms (SNPs) covering these four genes in 2044 Chinese Han subjects. Case-control analyses were performed to detect association of these 4 genes with schizophrenia and multifactor dimensionality reduction (MDR) analysis was employed to examine their potential gene-gene interaction in schizophrenia. Neither allelic nor genotypic single-locus tests revealed any significant association between EGR1-4 and the risk of schizophrenia nor was any such association found with regard to interaction within EGR1-4 (p(min)=0.623, CV Consistency=10/10). We concluded that although multiple candidate genes are involved in schizophrenogenic development, the EGR family may not play a major role in schizophrenia susceptibility in the Chinese Han population.
Collapse
Affiliation(s)
- Bao-Cheng Liu
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Analysis of factors decreasing testis weight in MRL mice. Mamm Genome 2010; 21:153-61. [DOI: 10.1007/s00335-010-9251-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 01/21/2010] [Indexed: 10/19/2022]
|
31
|
Leclerc N, Noh T, Cogan J, Samarawickrama DB, Smith E, Frenkel B. Opposing effects of glucocorticoids and Wnt signaling on Krox20 and mineral deposition in osteoblast cultures. J Cell Biochem 2008; 103:1938-51. [DOI: 10.1002/jcb.21587] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
32
|
Georgiou I, Pardalidis N, Giannakis D, Saito M, Watanabe T, Tsounapi P, Loutradis D, Kanakas N, Karagiannis A, Baltogiannis D, Giotitsas N, Miyagawa I, Sofikitis N. In vitro spermatogenesis as a method to bypass pre-meiotic or post-meiotic barriers blocking the spermatogenetic process: genetic and epigenetic implications in assisted reproductive technology. Andrologia 2007; 39:159-76. [PMID: 17714214 DOI: 10.1111/j.1439-0272.2007.00778.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Pregnancies achieved by assisted reproduction technologies and particularly by ooplasmic injections of either in vivo or in vitro generated immature male germ cells are susceptible to genetic risks inherent to the male population treated with assisted reproduction and additional risks inherent to these innovative procedures. The documented, as well as the theoretical risks, are discussed in this review. These risks represent mainly the consequences of genetic abnormalities underlying male infertility and may become stimulators for the development of novel approaches and applications in the treatment of infertility. Recent data suggest that techniques employed for in vitro spermatogenesis, male somatic cell haploidization, stem cell differentiation in vitro and assisted reproductive technology may also affect the epigenetic characteristics of the male gamete, the female gamete, or may have an impact on early embryogenesis. They may be also associated with an increased risk for genomic imprinting abnormalities. Production of haploid male gametes in vitro may not allow the male gamete to undergo all the genetic and epigenetic alterations that the male gamete normally undergoes during in vivo spermatogenesis.
Collapse
Affiliation(s)
- I Georgiou
- Laboratory of Molecular Urology and Genetics of Human Reproduction, Department of Urology, Ioannina University School of Medicine, Ioannina, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lawson MA, Tsutsumi R, Zhang H, Talukdar I, Butler BK, Santos SJ, Mellon PL, Webster NJG. Pulse sensitivity of the luteinizing hormone beta promoter is determined by a negative feedback loop Involving early growth response-1 and Ngfi-A binding protein 1 and 2. Mol Endocrinol 2007; 21:1175-91. [PMID: 17299135 PMCID: PMC2932486 DOI: 10.1210/me.2006-0392] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The hypothalamic-pituitary-gonadal endocrine axis regulates reproduction through estrous phase-dependent release of the heterodimeric gonadotropic glycoprotein hormones, LH and FSH, from the gonadotropes of the anterior pituitary. Gonadotropin synthesis and release is dependent upon pulsatile stimulation by the hypothalamic neuropeptide GnRH. Alterations in pulse frequency and amplitude alter the relative levels of gonadotropin synthesis and release. The mechanism of interpretation of GnRH pulse frequency and amplitude by gonadotropes is not understood. We have examined gene expression in LbetaT2 gonadotropes under various pulse regimes in a cell perifusion system by microarray and identified 1127 genes activated by tonic or pulsatile GnRH. Distinct patterns of expression are associated with each pulse frequency, but the greatest changes occur at a 60-min or less interpulse interval. The immediate early gene mRNAs encoding early growth response (Egr)1 and Egr2, which activate the gonadotropin LH beta-subunit gene promoter, are stably induced at high pulse frequency. In contrast, mRNAs for the Egr corepressor genes Ngfi-A binding protein Nab1 and Nab2 are stably induced at low pulse frequency. We show that Ngfi-A binding protein members inhibit Egr-mediated frequency-dependent induction of the LH beta-subunit promoter. This pattern of expression suggests a model of pulse frequency detection that acts by suppressing activation by Egr family members at low frequency and allowing activation at sustained high-frequency pulses.
Collapse
Affiliation(s)
- Mark A Lawson
- Department of Reproductive Medicine 0674, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Georgiou I, Syrrou M, Pardalidis N, Karakitsios K, Mantzavinos T, Giotitsas N, Loutradis D, Dimitriadis F, Saito M, Miyagawa I, Tzoumis P, Sylakos A, Kanakas N, Moustakareas T, Baltogiannis D, Touloupides S, Giannakis D, Fatouros M, Sofikitis N. Genetic and epigenetic risks of intracytoplasmic sperm injection method. Asian J Androl 2007; 8:643-73. [PMID: 17111067 DOI: 10.1111/j.1745-7262.2006.00231.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pregnancies achieved by assisted reproduction technologies, particularly by intracytoplasmic sperm injection (ICSI) procedures, are susceptible to genetic risks inherent to the male population treated with ICSI and additional risks inherent to this innovative procedure. The documented, as well as the theoretical, risks are discussed in the present review study. These risks mainly represent that consequences of the genetic abnormalities underlying male subfertility (or infertility) and might become stimulators for the development of novel approaches and applications in the treatment of infertility. In addition, risks with a polygenic background appearing at birth as congenital anomalies and other theoretical or stochastic risks are discussed. Recent data suggest that assisted reproductive technology might also affect epigenetic characteristics of the male gamete, the female gamete, or might have an impact on early embryogenesis. It might be also associated with an increased risk for genomic imprinting abnormalities.
Collapse
Affiliation(s)
- Ioannis Georgiou
- Laboratory of Molecular Urology and Genetics of Human Reproduction, Department of Urology, Ioannina University School of Medicine, Ioannina 45110, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Baron V, Adamson ED, Calogero A, Ragona G, Mercola D. The transcription factor Egr1 is a direct regulator of multiple tumor suppressors including TGFbeta1, PTEN, p53, and fibronectin. Cancer Gene Ther 2006; 13:115-24. [PMID: 16138117 PMCID: PMC2455793 DOI: 10.1038/sj.cgt.7700896] [Citation(s) in RCA: 279] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent studies are reviewed indicating that the transcription factor early growth response-1 (Egr1) is a direct regulator of multiple tumor suppressors including TGFbeta1, PTEN, p53, and fibronectin. The downstream pathways of these factors display multiple nodes of interaction with each other, suggesting the existence of a functional network of suppressor factors that serve to maintain normal growth regulation and resist the emergence of transformed variants. Paradoxically, Egr1 is oncogenic in prostate cancer. In the majority of these cancers, PTEN or p53 is inactive. It is suggested that these defects in the suppressor network allow for the unopposed induction of TGFbeta1 and fibronectin, which favor transformation and survival of prostate tumor epithelial cells, and explain the role of Egr1 in prostate cancer. Egr1 is a novel and logical target for intervention by gene therapy methods, and targeting methods are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Dan Mercola
- The Burnham Institute, La Jolla, CA 92037
- The Rebecca and John Moores Cancer Center, University of California at San Diego, La Jolla, CA 92093
- The Department of Pathology, University of California at Irvine, Irvine, CA 92697
| |
Collapse
|
36
|
Bolor H, Wakasugi N, Zhao WD, Ishikawa A. Detection of quantitative trait loci causing abnormal spermatogenesis and reduced testis weight in the small testis (Smt) mutant mouse. Exp Anim 2006; 55:97-108. [PMID: 16651692 DOI: 10.1538/expanim.55.97] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The small testis (Smt) mutant mouse is characterized by a small testis of one third to one half the size of a normal testis, and its spermatogenesis is mostly arrested at early stages of meiosis, although a small number of spermatocytes at the late prophase of meiosis and a few spermatids can sometimes be seen. We performed quantitative trait locus (QTL) analysis of these spermatogenic traits and testis weight using 221 F2 males obtained from a cross between Smt and MOM (Mus musculus molossinus) mice. At the genome-wide 5% level, we detected two QTLs affecting meiosis on chromosomes 4 and 13, and two QTLs for paired testis weight as a percentage of body weight on chromosomes 4 and X. In addition, we found several QTLs for degenerated germ cells and multinuclear giant cells on chromosomes 4, 7 and 13. Interestingly, for cell degeneration, the QTL on chromosome 13 interacted epistatically with the QTL on chromosome 4. These results reveal polygenic participation in the abnormal spermatogenesis and small testis size in the Smt mutant.
Collapse
Affiliation(s)
- Hasbaira Bolor
- Division of Applied Genetics and Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | | | | | | |
Collapse
|
37
|
Zhang H, Bailey JS, Coss D, Lin B, Tsutsumi R, Lawson MA, Mellon PL, Webster NJG. Activin modulates the transcriptional response of LbetaT2 cells to gonadotropin-releasing hormone and alters cellular proliferation. Mol Endocrinol 2006; 20:2909-30. [PMID: 16772531 PMCID: PMC2673912 DOI: 10.1210/me.2006-0109] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Both GnRH and activin are crucial for the correct function of pituitary gonadotrope cells. GnRH regulates LH and FSH synthesis and secretion and gonadotrope proliferation, whereas activin is essential for expression of FSH. Little is known, however, about the interplay of signaling downstream of these two hormones. In this study, we undertook expression profiling to determine how activin pretreatment alters the transcriptional response of LbetaT2 gonadotrope cells to GnRH stimulation. Activin treatment alone altered the transcriptional profile of 303 genes including inducing that of the 17beta-hydroxysteroid dehydrogenase B1 gene that converts estrone to 17beta-estradiol, altering the sensitivity of the cells to estrone. Furthermore, activin had a dramatic effect on the response of LbetaT2 cells to GnRH. Hierarchical clustering of 2453 GnRH-responsive genes identified groups of genes the response of which to GnRH was either enhanced or blunted after activin treatment. Mapping of these genes to gene ontology classifications or signaling pathways highlighted significant differences in the classes of altered genes. In the presence of activin, GnRH regulates genes in pathways controlling cell energetics, cytoskeletal rearrangements, organelle organization, and mitosis in the absence of activin, but genes controlling protein processing, cell differentiation, and secretion. Therefore, we demonstrated that activin enhanced GnRH induction of p38MAPK activity, caused GnRH-dependent phosphorylation of p53, and reduced the ability of GnRH to cause G1 arrest. Thus, although activin alone changes a modest number of transcripts, activin pretreatment dramatically alters the response to GnRH from an antiproliferative response to a more differentiated, synthetic response appropriate for a secretory cell.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0673, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Yamada D, Yoshida M, Williams YN, Fukami T, Kikuchi S, Masuda M, Maruyama T, Ohta T, Nakae D, Maekawa A, Kitamura T, Murakami Y. Disruption of spermatogenic cell adhesion and male infertility in mice lacking TSLC1/IGSF4, an immunoglobulin superfamily cell adhesion molecule. Mol Cell Biol 2006; 26:3610-24. [PMID: 16612000 PMCID: PMC1447408 DOI: 10.1128/mcb.26.9.3610-3624.2006] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
TSLC1/IGSF4, an immunoglobulin superfamily molecule, is predominantly expressed in the brain, lungs, and testes and plays important roles in epithelial cell adhesion, cancer invasion, and synapse formation. We generated Tslc1/Igsf4-deficient mice by disrupting exon 1 of the gene and found that Tslc1(-/-) mice were born with the expected Mendelian ratio but that Tslc1(-/-) male mice were infertile. In 11-week-old adult Tslc1(-/-) mice, the weight of a testis was 88% that in Tslc1(+/+) mice, and the number of sperm in the semen was approximately 0.01% that in Tslc1(+/+) mice. Histological analysis revealed that the round spermatids and the pachytene spermatocytes failed to attach to the Sertoli cells in the seminiferous tubules and sloughed off into the lumen with apoptosis in the Tslc1(-/-) mice. On the other hand, the spermatogonia and the interstitial cells, including Leydig cells, were essentially unaffected. In the Tslc1(+/+) mice, TSLC1/IGSF4 expression was observed in the spermatogenic cells from the intermediate spermatogonia to the early pachytene spermatocytes and from spermatids at step 7 or later. These findings suggest that TSLC1/IGSF4 expression is indispensable for the adhesion of spermatocytes and spermatids to Sertoli cells and for their normal differentiation into mature spermatozoa.
Collapse
Affiliation(s)
- Daisuke Yamada
- Tumor Suppression and Functional Genomics Project, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Meiosis is a unique form of cellular division by which a diploid cell produces genetically distinct haploid gametes. Initiation and regulation of mammalian meiosis differs between the sexes. In females, meiosis is initiated during embryo development and arrested shortly after birth during prophase I. In males, spermatogonial stem cells initiate meiosis at puberty and proceed through gametogenesis with no cell cycle arrest. Mouse genes required for early meiotic cell cycle events are being identified by comparative analysis with other eukaryotic systems, by virtue of gene knockout technology and by mouse mutagenesis screens for reproductive defects. This review focuses on mouse reproductive biology and describes the available mouse mutants with defects in the early meiotic cell cycle and prophase I regulatory events. These research tools will permit rapid advances in such medically relevant research areas as infertility, embryo lethality and developmental abnormalities.
Collapse
Affiliation(s)
- Changanamkandath Rajesh
- Department of Physiology and Cardiovascular Genomics, Medical University of Ohio, Toledo 43614, USA
| | | |
Collapse
|
40
|
Sarateanu CS, Retuerto MA, Beckmann JT, McGregor L, Carbray J, Patejunas G, Nayak L, Milbrandt J, Rosengart TK. An Egr-1 master switch for arteriogenesis: Studies in Egr-1 homozygous negative and wild-type animals. J Thorac Cardiovasc Surg 2006; 131:138-45. [PMID: 16399305 DOI: 10.1016/j.jtcvs.2005.08.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 07/12/2005] [Accepted: 08/02/2005] [Indexed: 11/15/2022]
Abstract
BACKGROUND Arteriogenesis has been implicated as an important biologic response to acute vascular occlusion. The early growth response 1 (Egr-1) gene encodes an immediate-early response transcription factor that is upregulated by changes in vascular strain and that in turn upregulates a number of putative angiogenic and arteriogenic growth factors. We therefore hypothesized that early growth response 1 might be a critical arteriogenic messenger that induces revascularization in the setting of acute vascular occlusions. METHODS Wild-type or Egr-1-/- (null) C57 BL mice, or Sprague-Dawley rats, underwent unilateral iliofemoral artery excision and subsequent analyses for angiogenesis and arteriogenesis through cell-specific immunohistochemistry. Rats were also administered an adenoviral vector encoding for Egr-1 (AdEgr group), noncoding vectors (AdNull group), or saline, after which these animals were assessed by means of serial laser Doppler perfusion imaging and morphologic examination of rat foot-pad ischemic lesions. RESULTS Egr-1 wild-type mice demonstrated an equivalent number of capillaries but a greater number of arterioles following excision versus Egr-1 null mice. AdEgr group rats demonstrated greater distal perfusion from 7 to 21 days after excision compared with control animals (P < .02), which approximated normal perfusion at 21 days after excision. AdEgr group rats also demonstrated greater arteriolar density and less severe ischemic foot-pad lesions than control animals. CONCLUSION These data suggest the importance of Egr-1 as a critical and potentially therapeutic mediator of revascularization after vascular occlusion and implicate arteriogenesis (collateral vessel formation) as a critical component of this process.
Collapse
|
41
|
Li L, Carter J, Gao X, Whitehead J, Tourtellotte WG. The neuroplasticity-associated arc gene is a direct transcriptional target of early growth response (Egr) transcription factors. Mol Cell Biol 2005; 25:10286-300. [PMID: 16287845 PMCID: PMC1291244 DOI: 10.1128/mcb.25.23.10286-10300.2005] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Accepted: 09/02/2005] [Indexed: 11/20/2022] Open
Abstract
Early growth response (Egr) transcription factors (Egr1 to Egr4) are synaptic activity-inducible immediate early genes (IEGs) that regulate some aspects of synaptic plasticity-related to learning and memory, yet the target genes regulated by them are unknown. In particular, Egr1 is essential for persistence of late-phase long-term potentiation (L-LTP), for hippocampus-dependent long-term memory formation, and for reconsolidation of previously established memories. Here, we show that Egr1 and Egr3 directly regulate the plasticity-associated activity-regulated cytoskeletal-related (Arc) gene, a synaptic activity-induced effector molecule which is also required for L-LTP and hippocampus-dependent learning and memory processing. Moreover, Egr1-deficient and Egr3-deficient mice lack Arc protein in a subpopulation of neurons, while mice lacking both Egr1 and Egr3 lack Arc in all neurons. Thus, Egr1 and Egr3 can indirectly modulate synaptic plasticity by directly regulating Arc and the plasticity mechanisms it mediates in recently activated synapses.
Collapse
Affiliation(s)
- Lin Li
- Ward 7-110, Department of Pathology, W127, Northwestern University, 393 E. Chicago Ave., Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
42
|
Sofikitis N, Pappas E, Kawatani A, Baltogiannis D, Loutradis D, Kanakas N, Giannakis D, Dimitriadis F, Tsoukanelis K, Georgiou I, Makrydimas G, Mio Y, Tarlatzis V, Melekos M, Miyagawa I. Efforts to create an artificial testis: culture systems of male germ cells under biochemical conditions resembling the seminiferous tubular biochemical environment. Hum Reprod Update 2005; 11:229-59. [PMID: 15817525 DOI: 10.1093/humupd/dmi007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Induction of meiotic and post-meiotic alterations of male germ cells in vitro has been the target of several research efforts since 1960. However, to date, the establishment of an ideal culture system in which spermatogonial stem cells can be maintained and directed to proliferate and undergo meiosis and complete spermiogenesis does not exist. This is attributed to the difficulties concerning the isolation and purification of defined subpopulations of germ cells and the establishment of male germ cell lines. In addition, there is no adequate knowledge regarding the optimal biochemical conditions that promote the survival and differentiation of germ cells in long-term cultures. This review focuses on the methodologies that have been proved sufficient to achieve differentiation of cultured male germ cells. Furthermore, the factors regulating spermatogenesis and the technical prerequisites to achieve differentiation of cultured male germ cells are described. Finally, the role of in vitro cultures of immature diploid germ cells in the therapeutic management of men negative for haploid cells in their testes and the subsequent potential genetic and epigenetic risks are discussed.
Collapse
Affiliation(s)
- N Sofikitis
- Laboratory for Molecular Urology and Genetics of Human Reproduction, Department of Urology, Ioannina University School of Medicine, Ioannina, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Nakamoto T, Shiratsuchi A, Oda H, Inoue K, Matsumura T, Ichikawa M, Saito T, Seo S, Maki K, Asai T, Suzuki T, Hangaishi A, Yamagata T, Aizawa S, Noda M, Nakanishi Y, Hirai H. Impaired spermatogenesis and male fertility defects in CIZ/Nmp4-disrupted mice. Genes Cells 2005; 9:575-89. [PMID: 15189450 DOI: 10.1111/j.1356-9597.2004.00746.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CIZ (Cas interacting zinc finger protein), also called Nmp4 (nuclear matrix protein 4), is a nucleo-cytoplasmic shuttling transcription factor that regulates the expression of collagen and matrix metalloproteinases. CIZ/Nmp4 was originally cloned by its binding to p130(Cas), a focal adhesion protein, and was recently shown to suppress BMP2 (bone mophogenetic protein 2) signalling. To explore the physiological role of CIZ/Nmp4, we disrupted CIZ/Nmp4-gene by inserting beta-galactosidase and neomycin resistance genes into the 2nd exon of CIZ/Nmp4-gene, which is utilized by all the sequenced alternative forms. CIZ-/- mice were born and grew to adulthood. Although they tend to be smaller than wild-type mice, no pathological abnormality was observed except in the testis. Histological analysis of the testes revealed variable degrees of spermatogenic cell degeneration within the seminiferous tubules of CIZ-/- mice, resembling the histology of the 'Germinal-cell aplasia with focal spermatogenesis'. Some of the CIZ-/- male mice developed infertility. TUNEL assay on testis sections revealed an increased occurrence of apoptosis of spermatogenic cells in the testes of CIZ-/- mice. CIZ/Nmp4 was co-localized with Smad1 in the testis, suggesting that a disregulation of BMP signalling could cause these phenotypes. These results suggest that CIZ/Nmp4 plays roles in the progress and the maintenance of spermatogenesis.
Collapse
Affiliation(s)
- Tetsuya Nakamoto
- Department of Haematology & Oncology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Good JM, Nachman MW. Rates of Protein Evolution Are Positively Correlated with Developmental Timing of Expression During Mouse Spermatogenesis. Mol Biol Evol 2005; 22:1044-52. [PMID: 15647515 DOI: 10.1093/molbev/msi087] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Male reproductive genes often evolve very rapidly, and sexual selection is thought to be a primary force driving this divergence. We investigated the molecular evolution of 987 genes expressed at different times during mouse spermatogenesis to determine if the rate of evolution and the intensity of positive selection vary across stages of male gamete development. Using mouse-rat orthologs, we found that rates of protein evolution were positively correlated with the developmental timing of expression. Genes expressed early in spermatogenesis had rates of divergence similar to the genome median, while genes expressed after the onset of meiosis were found to evolve much more quickly. Rates of protein evolution were fastest for genes expressed during the dramatic morphogenesis of round spermatids into spermatozoa. Late-expressed genes were also more likely to be specific to the male germline. To test for evidence of positive selection, we analyzed the ratio of nonsynonymous to synonymous changes using a maximum likelihood framework in comparisons among mouse, rat, and human. Many genes showed evidence of positive selection, and most of these genes were expressed late in spermatogenesis and were testis specific. Overall, these data suggest that the intensity of positive selection associated with the evolution of male gametes varies considerably across development and acts primarily on phenotypes that develop late in spermatogenesis.
Collapse
Affiliation(s)
- Jeffrey M Good
- Department of Ecology and Evolutionary Biology, University of Arizona, USA.
| | | |
Collapse
|
45
|
Hagihara A, Miyamoto K, Furuta J, Hiraoka N, Wakazono K, Seki S, Fukushima S, Tsao MS, Sugimura T, Ushijima T. Identification of 27 5' CpG islands aberrantly methylated and 13 genes silenced in human pancreatic cancers. Oncogene 2004; 23:8705-10. [PMID: 15467763 DOI: 10.1038/sj.onc.1207783] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Aberrantly methylated DNA fragments were searched for in human pancreatic cancers, using the genome scanning technique: methylation-sensitive-representational difference analysis (MS-RDA). MS-RDA isolated 111 DNA fragments derived from CpG islands (CGIs), and 35 of them were from CGIs in the 5' regions of known genes. Methylation-specific PCR (MSP) of the CGIs in seven pancreatic cancer cell lines and two pancreatic ductal epithelial cell lines showed that 27 CGIs in the 5' regions were aberrantly methylated in at least one of the cancer cell lines. Quantitative reverse-transcription-PCR analysis showed that downstream genes of all the CGIs were either not expressed or only very weakly expressed in cancer cell lines with the aberrant methylation. In the pancreatic ductal epithelial cell lines, 18 genes were expressed at various levels, and nine genes were not expressed at all. Treatment of a cancer cell line with a demethylating agent, 5-aza-2'-deoxycytidine, restored the expression of 13 genes, RASGRF2, ADAM23, NEF3, NKX2-8, HAND1, EGR4, PRG2, FBN2, CDH2, TLL1, NPTX1, NTSR1 and THBD, showing their silencing by methylation of their 5' CGIs. MSP of 24 primary pancreatic cancers showed that all these genes, except for THBD, were methylated in at least one cancer. Some of those were suggested to be potentially involved in pancreatic cancer development and progression.
Collapse
Affiliation(s)
- Atsushi Hagihara
- Carcinogenesis Division, National Cancer Center Research Institute, 1-1 Tsukiji 5-chome, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Schalch P, Patejunas G, Retuerto M, Sarateanu S, Milbrandt J, Thakker G, Kim D, Carbray J, Crystal RG, Rosengart TK. Homozygous deletion of early growth response 1 gene and critical limb ischemia after vascular ligation in mice: evidence for a central role in vascular homeostasis. J Thorac Cardiovasc Surg 2004; 128:595-601. [PMID: 15457161 DOI: 10.1016/j.jtcvs.2004.02.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The early growth response 1 gene (Egr1) encodes for an immediate to early response transcription factor that is upregulated by changes in vascular strain and hypoxia and in turn upregulates the downstream expressions of a number of angiogenic growth factors. We therefore hypothesized that early growth response 1 may be a critical early messenger governing revascularization in the setting of acute vascular occlusions. METHODS C57 BL/6 mice deficient in the Egr1 gene (knockout) and their wild-type litter mates underwent ligation and excision of the femoral artery with or without the previous administration of 2.7 x 10(9) particle units of an adenoviral vector coding for the vascular endothelial growth factor gene (VEGF) or Egr1. Distal hind limb perfusion was serially measured in these animals with laser Doppler perfusion imaging. RESULTS Wild-type mice (n = 9) had nearly complete restitution of hind limb perfusion by day 35 after ligation. In contrast, all noninjected Egr1 knockout mice (n = 5) had severe ipsilateral limb necrosis develop between 1 and 4 days after ligation (P <.0001). Egr1 knockout mice injected with VEGF vector (n = 4) demonstrated significantly improved perfusion relative to baseline by postligation day 28, which persisted to postligation day 35 (P <.05). Egr1 knockout animals injected with Egr1 vector (n = 7) demonstrated a partial recovery of hind limb perfusion relative to VEGF vector-treated knockout animals at postligation day 4 (P <.01), which persisted to day 35. CONCLUSIONS These findings suggest that early growth response 1 plays a pivotal role in reperfusion responses to vascular occlusion in mice and possibly other mammals.
Collapse
Affiliation(s)
- Paul Schalch
- Evanston Northwestern Healthcare, Evanston, Ill., USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Jorgensen JS, Quirk CC, Nilson JH. Multiple and overlapping combinatorial codes orchestrate hormonal responsiveness and dictate cell-specific expression of the genes encoding luteinizing hormone. Endocr Rev 2004; 25:521-42. [PMID: 15294880 DOI: 10.1210/er.2003-0029] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Normal reproductive function in mammals requires precise control of LH synthesis and secretion by gonadotropes of the anterior pituitary. Synthesis of LH requires expression of two genes [alpha-glycoprotein subunit (alphaGSU) and LHbeta] located on different chromosomes. Hormones from the hypothalamus and gonads modulate transcription of both genes as well as secretion of the biologically active LH heterodimer. In males and females, the transcriptional tone of the genes encoding alphaGSU and LHbeta reflects dynamic integration of a positive signal provided by GnRH from hypothalamic neurons and negative signals emanating from gonadal steroids. Although alphaGSU and LHbeta genes respond transcriptionally in the same manner to changes in hormonal input, different combinations of regulatory elements orchestrate their response. These hormone-responsive regulatory elements are also integral members of much larger combinatorial codes responsible for targeting expression of alphaGSU and LHbeta genes to gonadotropes. In this review, we will profile the genomic landscape of the promoter-regulatory region of both genes, depicting elements and factors that contribute to gonadotrope-specific expression and hormonal regulation. Within this context, we will highlight the different combinatorial codes that control transcriptional responses, particularly those that mediate the opposing effects of GnRH and one of the sex steroids, androgens. We will use this framework to suggest that GnRH and androgens attain the same transcriptional endpoint through combinatorial codes unique to alphaGSU and LHbeta. This parallelism permits the dynamic and coordinate regulation of two genes that encode a single hormone.
Collapse
Affiliation(s)
- Joan S Jorgensen
- Department of Veterinary Biosciences, University of Illinois, Urbana 61802, USA
| | | | | |
Collapse
|
48
|
Hamra FK, Schultz N, Chapman KM, Grellhesl DM, Cronkhite JT, Hammer RE, Garbers DL. Defining the spermatogonial stem cell. Dev Biol 2004; 269:393-410. [PMID: 15110708 DOI: 10.1016/j.ydbio.2004.01.027] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2003] [Revised: 01/21/2004] [Accepted: 01/23/2004] [Indexed: 12/22/2022]
Abstract
Through the use of donor cells from transgenic rats expressing GFP exclusively in the germline, we have defined culture conditions where male germ cells lose (on STO cells) or maintain (on MSC-1 cells) stem cell activity. A cadre of germ cell transcripts strikingly decrease in relative abundance as a function of testis age or culture time on STO cells, but only a subset of these transcripts (approximately 248) remain elevated when cultured on MSC-1 cells. If specific gene expression regulates stem cell activity, some or all of these transcripts are candidates as such regulators. We establish a spermatogonial stem cell index (SSCI) that reliably predicts relative stem cell activity in rat or mouse testis cell cultures, and through the use of an antibody to a robust signal (Egr3) within the index find intense signals in single or paired cells. As germ cells form longer interconnected chains (incomplete cytokinesis), the Egr3 signal disappears coincident with a loss of stem cell activity. Thus, molecular markers specific for spermatogonial stem cells establish a reliable and rapid means by which to define these cells in culture and alleviate the need for laborious testicular transfers in initial cell culture studies.
Collapse
Affiliation(s)
- F Kent Hamra
- Cecil H and H Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Nissen PH, Thomsen B, Offenberg H, Thomsen PD, Bendixen C. Cloning and characterization of the bovine EGR4 gene and evaluation as candidate gene for bovine spinal dysmyelination. Anim Genet 2003; 34:124-31. [PMID: 12648095 DOI: 10.1046/j.1365-2052.2003.00969.x] [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: 11/20/2022]
Abstract
Genes of the early growth response (EGR) family encode transcription factors with a highly conserved DNA binding zinc finger domain, which regulate a variety of genes, e.g. late myelin genes. Here, the cloning, genomic structure and expression of the bovine orthologue of the EGR4 gene are reported. The gene consists of two exons and encodes a 482 amino acid protein with a Cys2His2 zinc finger structure. The predicted protein shares between 80 and 87% identity to mouse, rat and human EGR4 proteins and all four species share almost complete identity in the DNA-binding domain. The bovine transcript is alternatively spliced by retaining intronic sequence, giving rise to two different mRNAs differing in three nucleotides and resulting in an extra alanine residue in the longer variant of the predicted protein. The gene was mapped by radiation hybrid (RH) mapping to markers on bovine chromosome 11. EGR4 transcripts were detected by reverse transcriptase polymerase chain reaction (RT-PCR) in the frontal cortex and cerebellum, and a low expression level was also detected in the liver. The EGR4 gene was evaluated as a candidate gene for bovine spinal dysmyelination (BSD). Sequencing of the gene from a homozygous affected animal and a heterozygous carrier revealed a single base mutation that leads to an amino acid substitution at residue 322 in EGR4. Genotype analysis of this polymorphism in a pedigree segregating for BSD, as well as in a panel of different cattle breeds, and sequence analysis of the entire coding region suggested that the EGR4 is not the gene responsible for BSD. Furthermore, 87 animals of different cattle breeds were screened for single-nucleotide polymorphisms (SNPs) resulting in the identification of two SNPs in EGR4.
Collapse
Affiliation(s)
- P H Nissen
- Department of Animal Breeding and Genetics, Danish Institute of Agricultural Sciences, DK-8830 Tjele, Denmark.
| | | | | | | | | |
Collapse
|
50
|
Abstract
Spermatogenesis is a complex process that involves stem-cell renewal, genome reorganization and genome repackaging, and that culminates in the production of motile gametes. Problems at all stages of spermatogenesis contribute to human infertility, but few of them can be modelled in vitro or in cell culture. Targeted mutagenesis in the mouse provides a powerful method to analyse these steps and has provided new insights into the origins of male infertility.
Collapse
Affiliation(s)
- Howard J Cooke
- MRC Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK.
| | | |
Collapse
|