1
|
Tsinopoulou VR, Bacopoulou F, Fidani S, Christoforidis A. Genetic determinants of age at menarche: does the LIN28B gene play a role? A narrative review. Hormones (Athens) 2024:10.1007/s42000-024-00594-3. [PMID: 39227549 DOI: 10.1007/s42000-024-00594-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024]
Abstract
Menarche, the first menstrual period marking the onset of female reproduction, is a milestone of female puberty. The timing of menarche determines the timing of later phases of pubertal maturation in girls and has major implications for health later in life, including behavioral and psychosocial disorders during adolescence and fertility problems and increased risk for certain diseases in adulthood. Over the last few decades, a continuous decline in age at menarche has been noted, with environmental factors contributing to this change in the timing of menarche. However, a genetic component of age at menarche and pubertal onset has been strongly suggested by studies in families and twins wherein up to approximately 80% of the variance in puberty onset can be explained by heritability. Gene association studies have revealed several genetic loci involved in age at menarche, among which LIN28B has emerged as a key regulator of female growth and puberty. LIN28B, a human homolog of Lin28 of C. elegans, is a known RNA-binding protein that regulates let-7 microRNA biogenesis. Genome-wide association studies have identified the association of polymorphisms in the LIN28B gene with age at menarche in several population cohorts worldwide. In this paper, we review the genetic factors contributing to age of menarche, with particular focus on the identified polymorphisms in LIN28B gene.
Collapse
Affiliation(s)
- Vasiliki Rengina Tsinopoulou
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, University General Hospital AHEPA, Stilponos Kyriakidi 1, Thessaloniki, 54636, Greece.
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Athens, Greece
| | - Styliani Fidani
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, University General Hospital AHEPA, Stilponos Kyriakidi 1, Thessaloniki, 54636, Greece
- Laboratory of Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Christoforidis
- 1st Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Ippokratio General Hospital, Thessaloniki, Greece
| |
Collapse
|
2
|
Li Q, Chao T, Wang Y, Xuan R, Guo Y, He P, Zhang L, Wang J. Transcriptome analysis reveals miRNA expression profiles in hypothalamus tissues during the sexual development of Jining grey goats. BMC Genomics 2024; 25:832. [PMID: 39232653 PMCID: PMC11373458 DOI: 10.1186/s12864-024-10735-y] [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: 03/06/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Exploring the physiological and molecular mechanisms underlying goat sexual maturation can enhance breeding practices and optimize reproductive efficiency and is therefore substantially important for practical breeding purposes. As an essential neuroendocrine organ in animals, the hypothalamus is involved in sexual development and other reproductive processes in female animals. Although microRNAs (miRNAs) have been identified as significant regulators of goat reproduction, there is a lack of research on the molecular regulatory mechanisms of hypothalamic miRNAs that are involved in the sexual development of goats. Therefore, we examined the dynamic changes in serum hormone profiles and hypothalamic miRNA expression profiles at four developmental stages (1 day (neonatal, D1, n = 5), 2 months (prepubertal, M2, n = 5), 4 months (sexual maturity, M4, n = 5), and 6 months (breeding period, M6, n = 5)) during sexual development in Jining grey goats. RESULTS Transcriptome analysis revealed 95 differentially expressed miRNAs (DEMs) in the hypothalamus of goats across the four developmental stages. The target genes of these miRNAs were significantly enriched in the GnRH signalling pathway, the PI3K-Akt signalling pathway, and the Ras signalling pathway (P < 0.05). Additionally, 16 DEMs are common among the M2 vs. D1, M4 vs. D1, and M6 vs. D1 comparisons, indicating that the transition from D1 to M2 represents a potentially critical period for sexual development in Jining grey goats. The bioinformatics analysis results indicate that miR-193a/miR-193b-3p-Annexin A7 (ANXA7), miR-324-5p-Adhesion G protein-coupled receptor A1 (ADGRA1), miR-324-3p-Erbb2 receptor tyrosine kinase 2 (ERBB2), and miR-324-3p-Rap guanine nucleotide exchange factor 3 (RAPGEF3) are potentially involved in biological processes such as hormone secretion, energy metabolism, and signal transduction. In addition, we further confirmed that miR-324-3p targets the regulatory gene RAPGEF3. CONCLUSION These results further enrich the expression profile of hypothalamic miRNAs in goats and provide important insights for studying the regulatory effects of hypothalamic miRNAs on the sexual development of goats after birth.
Collapse
Affiliation(s)
- Qing Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Yanyan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Rong Xuan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Yanfei Guo
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Peipei He
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Lu Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China.
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Tai'an City, 271014, Shandong Province, China.
| |
Collapse
|
3
|
Marino M, D’Auria R, Mele E, Pastorino GMG, Di Pietro P, D’Angelo S, Della Rocca N, Operto FF, Vecchione C, Fasano S, Pierantoni R, Viggiano A, Meccariello R, Santoro A. The interplay between kisspeptin and endocannabinoid systems modulates male hypothalamic and gonadic control of reproduction in vivo. Front Endocrinol (Lausanne) 2023; 14:1269334. [PMID: 37900144 PMCID: PMC10602894 DOI: 10.3389/fendo.2023.1269334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/07/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Male reproduction is under the control of the hypothalamus-pituitary-gonadal (HPG) axis. The endocannabinoid system (ECS) and the kisspeptin system (KS) are two major signaling systems in the central and peripheral control of reproduction, but their possible interaction has been poorly investigated in mammals. This manuscript analyzes their possible reciprocal modulation in the control of the HPG axis. Materials and methods Adolescent male rats were treated with kisspeptin-10 (Kp10) and endocannabinoid anandamide (AEA), the latter alone or in combination with the type 1 cannabinoid receptor (CB1) antagonist rimonabant (SR141716A). The hypothalamic KS system and GnRH expression, circulating sex steroids and kisspeptin (Kiss1) levels, and intratesticular KS and ECS were evaluated by immunohistochemical and molecular methods. Non-coding RNAs (i.e., miR145-5p, miR-132-3p, let7a-5p, let7b-5p) were also considered. Results Circulating hormonal values were not significantly affected by Kp10 or AEA; in the hypothalamus, Kp10 significantly increased GnRH mRNA and aromatase Cyp19, Kiss1, and Kiss1 receptor (Kiss1R) proteins. By contrast, AEA treatment affected the hypothalamic KS at the protein levels, with opposite effects on the ligand and receptor, and SR141716A was capable of attenuating the AEA effects. Among the considered non-coding RNA, only the expression of miR145-5p was positively affected by AEA but not by Kp10 treatment. Localization of Kiss1+/Kiss1R+ neurons in the arcuate nucleus revealed an increase of Kiss1R-expressing neurons in Kp10- and AEA-treated animals associated with enlargement of the lateral ventricles in Kp10-treated animals. In the brain and testis, the selected non-coding RNA was differently modulated by Kp10 or AEA. Lastly, in the testis, AEA treatment affected the KS at the protein levels, whereas Kp10 affected the intragonadal levels of CB1 and FAAH, the main modulator of the AEA tone. Changes in pubertal transition-related miRNAs and the intratesticular distribution of Kiss1, Kiss1R, CB1, and CB2 following KP and AEA treatment corroborate the KS-ECS crosstalk also showing that the CB1 receptor is involved in this interplay. Conclusion For the first time in mammals, we report the modulation of the KS in both the hypothalamus and testis by AEA and revealed the KP-dependent modulation of CB1 and FAAH in the testis. KP involvement in the progression of spermatogenesis is also suggested.
Collapse
Affiliation(s)
- Marianna Marino
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| | - Raffaella D’Auria
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| | - Elena Mele
- Dipartimento di Scienze Motorie e del Benessere, Università di Napoli Parthenope, Napoli, Italy
| | - Grazia Maria Giovanna Pastorino
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
- Unità Operativa Complessa (U.O.C.) Neuropsichiatria Infantile, Azienda Ospedaliero Universitaria San Giovanni di Dio Ruggi d’Aragona, “Scuola Medica Salernitana”, Salerno, Italy
| | - Paola Di Pietro
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| | - Stefania D’Angelo
- Dipartimento di Scienze Motorie e del Benessere, Università di Napoli Parthenope, Napoli, Italy
| | - Natalia Della Rocca
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| | | | - Carmine Vecchione
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| | - Silvia Fasano
- Dipartimento di Medicina Sperimentale, Università della Campania L. Vanvitelli, Napoli, Italy
| | - Riccardo Pierantoni
- Dipartimento di Medicina Sperimentale, Università della Campania L. Vanvitelli, Napoli, Italy
| | - Andrea Viggiano
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| | - Rosaria Meccariello
- Dipartimento di Scienze Motorie e del Benessere, Università di Napoli Parthenope, Napoli, Italy
| | - Antonietta Santoro
- Dipartimento di Medicina, Chirurgia e Odontoiatria “Scuola Medica Salernitana” Università di Salerno, Baronissi, Italy
| |
Collapse
|
4
|
Xiao S, Zhang W, Li J, Manley NR. Lin28 regulates thymic growth and involution and correlates with MHCII expression in thymic epithelial cells. Front Immunol 2023; 14:1261081. [PMID: 37868985 PMCID: PMC10588642 DOI: 10.3389/fimmu.2023.1261081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 10/24/2023] Open
Abstract
Thymic epithelial cells (TECs) are essential for T cell development in the thymus, yet the mechanisms governing their differentiation are not well understood. Lin28, known for its roles in embryonic development, stem cell pluripotency, and regulating cell proliferation and differentiation, is expressed in endodermal epithelial cells during embryogenesis and persists in adult epithelia, implying postnatal functions. However, the detailed expression and function of Lin28 in TECs remain unknown. In this study, we examined the expression patterns of Lin28 and its target Let-7g in fetal and postnatal TECs and discovered opposing expression patterns during postnatal thymic growth, which correlated with FOXN1 and MHCII expression. Specifically, Lin28b showed high expression in MHCIIhi TECs, whereas Let-7g was expressed in MHCIIlo TECs. Deletion of Lin28a and Lin28b specifically in TECs resulted in reduced MHCII expression and overall TEC numbers. Conversely, overexpression of Lin28a increased total TEC and thymocyte numbers by promoting the proliferation of MHCIIlo TECs. Additionally, our data strongly suggest that Lin28 and Let-7g expression is reliant on FOXN1 to some extent. These findings suggest a critical role for Lin28 in regulating the development and differentiation of TECs by modulating MHCII expression and TEC proliferation throughout thymic ontogeny and involution. Our study provides insights into the mechanisms underlying TEC differentiation and highlights the significance of Lin28 in orchestrating these processes.
Collapse
Affiliation(s)
- Shiyun Xiao
- Department of Genetics, University of Georgia, Athens, GA, United States
| | | | | | | |
Collapse
|
5
|
Pereira SA, Oliveira FCB, Naulé L, Royer C, Neves FAR, Abreu AP, Carroll RS, Kaiser UB, Coelho MS, Lofrano-Porto A. Mouse Testicular Mkrn3 Expression Is Primarily Interstitial, Increases Peripubertally, and Is Responsive to LH/hCG. Endocrinology 2023; 164:bqad123. [PMID: 37585624 PMCID: PMC10449413 DOI: 10.1210/endocr/bqad123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Studies in humans and mice support a role for Makorin RING finger protein 3 (MKRN3) as an inhibitor of gonadotropin-releasing hormone (GnRH) secretion prepubertally, and its loss of function is the most common genetic cause of central precocious puberty in humans. Studies have shown that the gonads can synthesize neuropeptides and express MKRN3/Mkrn3 mRNA. Therefore, we aimed to investigate the spatiotemporal expression pattern of Mkrn3 in gonads during sexual development, and its potential regulation in the functional testicular compartments by gonadotropins. Mkrn3 mRNA was detected in testes and ovaries of wild-type mice at all ages evaluated, with a sexually dimorphic expression pattern between male and female gonads. Mkrn3 expression was highest peripubertally in the testes, whereas it was lower peripubertally than prepubertally in the ovaries. Mkrn3 is expressed primarily in the interstitial compartment of the testes but was also detected at low levels in the seminiferous tubules. In vitro studies demonstrated that Mkrn3 mRNA levels increased in human chorionic gonadotropin (hCG)-treated Leydig cell primary cultures. Acute administration of a GnRH agonist in adult mice increased Mkrn3 expression in testes, whereas inhibition of the hypothalamic-pituitary-gonadal axis by chronic administration of GnRH agonist had the opposite effect. Finally, we found that hCG increased Mkrn3 mRNA levels in a dose-dependent manner. Taken together, our developmental expression analyses, in vitro and in vivo studies show that Mkrn3 is expressed in the testes, predominantly in the interstitial compartment, and that Mkrn3 expression increases after puberty and is responsive to luteinizing hormone/hCG stimulation.
Collapse
Affiliation(s)
- Sidney A Pereira
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fernanda C B Oliveira
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Lydie Naulé
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carine Royer
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Francisco A R Neves
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Ana Paula Abreu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michella S Coelho
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
| | - Adriana Lofrano-Porto
- Molecular Pharmacology Laboratory, School of Health Sciences, University of Brasilia, Brasilia-DF, Brazil
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Zhao X, Sang M, Han P, Gao J, Liu Z, Li H, Gu Y, Wang C, Sun F. Peptides from the croceine croaker ( Larimichthys crocea) swim bladder attenuate busulfan-induced oligoasthenospermia in mice. PHARMACEUTICAL BIOLOGY 2022; 60:319-325. [PMID: 35148224 PMCID: PMC8843205 DOI: 10.1080/13880209.2022.2034895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/03/2022] [Accepted: 01/23/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT The swim bladder of the croceine croaker is believed to have a therapeutic effect on various diseases. However, there is no research about its effect on mammalian spermatogenesis. OBJECTIVE We investigated the swim bladder peptides (SBPs) effect on busulfan-induced oligoasthenospermia in mice. MATERIALS AND METHODS We first extracted SBP from protein hydrolysate of the croceine croaker swim bladder, and then five groups of ICR male mice were randomly assigned: control, control + SBP 60 mg/kg, busulfan, busulfan + SBP 30 mg/kg and busulfan + SBP 60 mg/kg. Mice received bilateral intratesticular injections of busulfan to establish oligoasthenospermia model. After treatment with SBP for 4 weeks, testis and epididymis were collected from all mice for further analysis. RESULTS After treatment with SBP 30-60 mg/kg for 4 weeks, epididymal sperm concentration and motility increased by 3.9-9.6- and 1.9-2.4-fold than those of oligoasthenospermia mice induced by busulfan. Meanwhile, histology showed that spermatogenic cells decreased, leading to increased lumen diameters and vacuolization in the busulfan group. These features were reversed by SBP treatment. RNA-sequencing analysis revealed that, compared with the busulfan group, Lin28b and Igf2bp1 expression related to germ cell proliferation, increased with a >1.5-fold change after SBP treatment. Additionally, PGK2 and Cfap69 mRNAs associated with sperm motility, also increased with a >1.5-fold change. Furthermore, these findings were validated by quantitative real-time PCR and Western blotting. DISCUSSION AND CONCLUSIONS This is the first reported evidence for the therapeutic effect of SBP on oligoasthenospermia. SBP may be a promising drug for oligoasthenospermia in humans.
Collapse
Affiliation(s)
- Xi Zhao
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Mengmeng Sang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Ping Han
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Jie Gao
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Zhenhua Liu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Hu Li
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Yayun Gu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Chengniu Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Sun
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
7
|
LIN28 Family in Testis: Control of Cell Renewal, Maturation, Fertility and Aging. Int J Mol Sci 2022; 23:ijms23137245. [PMID: 35806250 PMCID: PMC9266904 DOI: 10.3390/ijms23137245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 12/12/2022] Open
Abstract
Male reproductive development starts early in the embryogenesis with somatic and germ cell differentiation in the testis. The LIN28 family of RNA-binding proteins promoting pluripotency has two members—LIN28A and LIN28B. Their function in the testis has been investigated but many questions about their exact role based on the expression patterns remain unclear. LIN28 expression is detected in the gonocytes and the migrating, mitotically active germ cells of the fetal testis. Postnatal expression of LIN28 A and B showed differential expression, with LIN28A expressed in the undifferentiated spermatogonia and LIN28B in the elongating spermatids and Leydig cells. LIN28 interferes with many signaling pathways, leading to cell proliferation, and it is involved in important testicular physiological processes, such as cell renewal, maturation, fertility, and aging. In addition, aberrant LIN28 expression is associated with testicular cancer and testicular disorders, such as hypogonadotropic hypogonadism and Klinefelter’s syndrome. This comprehensive review encompasses current knowledge of the function of LIN28 paralogs in testis and other tissues and cells because many studies suggest LIN28AB as a promising target for developing novel therapeutic agents.
Collapse
|
8
|
Lin NW, Liu C, Yang IV, Maier LA, DeMeo DL, Wood C, Ye S, Cruse MH, Smith VL, Vyhlidal CA, Kechris K, Sharma S. Sex-Specific Differences in MicroRNA Expression During Human Fetal Lung Development. Front Genet 2022; 13:762834. [PMID: 35480332 PMCID: PMC9037032 DOI: 10.3389/fgene.2022.762834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/05/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Sex-specific differences in fetal lung maturation have been well described; however, little is known about the sex-specific differences in microRNA (miRNA) expression during human fetal lung development. Interestingly, many adult chronic lung diseases also demonstrate sex-specific differences in prevalence. The developmental origins of health and disease hypothesis suggests that these sex-specific differences in fetal lung development may influence disease susceptibility later in life. In this study, we performed miRNA sequencing on human fetal lung tissue samples to investigate differential expression of miRNAs between males and females in the pseudoglandular stage of lung development. We hypothesized that differences in miRNA expression are present between sexes in early human lung development and may contribute to the sex-specific differences seen in pulmonary diseases later in life. Methods: RNA was isolated from human fetal lung tissue samples for miRNA sequencing. The count of each miRNA was modeled by sex using negative binomial regression models in DESeq2, adjusting for post-conception age, age2, smoke exposure, batch, and RUV factors. We tested for differential expression of miRNAs by sex, and for the presence of sex-by-age interactions to determine if miRNA expression levels by age were distinct between males and females. Results: miRNA expression profiles were generated on 298 samples (166 males and 132 females). Of the 809 miRNAs expressed in human fetal lung tissue during the pseudoglandular stage of lung development, we identified 93 autosomal miRNAs that were significantly differentially expressed by sex and 129 miRNAs with a sex-specific pattern of miRNA expression across the course of the pseudoglandular period. Conclusion: Our study demonstrates differential expression of numerous autosomal miRNAs between the male and female developing human lung. Additionally, the expression of some miRNAs are modified by age across the pseudoglandular stage in a sex-specific way. Some of these differences in miRNA expression may impact susceptibility to pulmonary disease later in life. Our results suggest that sex-specific miRNA expression during human lung development may be a potential mechanism to explain sex-specific differences in lung development and may impact subsequent disease susceptibility.
Collapse
Affiliation(s)
- Nancy W. Lin
- Division of Environmental and Occupational Health, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Cuining Liu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Ivana V. Yang
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lisa A. Maier
- Division of Environmental and Occupational Health, National Jewish Health, Denver, CO, United States
- Environmental and Occupational Health, Colorado School of Public Health, Aurora, CO, United States
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Cheyret Wood
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Shuyu Ye
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Margaret H. Cruse
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Vong L. Smith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | | | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
9
|
Di C, Zheng G, Zhang Y, Tong E, Ren Y, Hong Y, Song Y, Chen R, Tan X, Yang L. RTA and LANA Competitively Regulate let-7a/RBPJ Signal to Control KSHV Replication. Front Microbiol 2022; 12:804215. [PMID: 35069510 PMCID: PMC8777081 DOI: 10.3389/fmicb.2021.804215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/03/2021] [Indexed: 11/13/2022] Open
Abstract
The recombination signal binding protein for immunoglobulin kappa J region (RBPJ) has a dual effect on Kaposi's sarcoma-associated herpesvirus (KSHV) replication. RBPJ interaction with replication and transcription activator (RTA) is essential for lytic replication, while the interaction with latency-associated nuclear antigen (LANA) facilitates latent infection. Furthermore, our previous study found that LANA decreased RBPJ through upregulating miRNA let-7a. However, it is unclear whether RTA regulates the expression of RBPJ. Here, we show RTA increases RBPJ by decreasing let-7a. During KSHV replication, the RBPJ expression level was positively correlated with the RTA expression level and negatively correlated with the LANA expression level. The let-7a expression level was inverse to RBPJ. Knockdown of RBPJ inhibited the self-activation of RTA promoter and LANA promoter and weakened LANA's inhibition of RTA promoter. Collectively, these findings indicate that RTA and LANA compete for let-7a/RBPJ signal to control the KSHV replication. Regulating the RBPJ expression level by RTA and LANA plays an important role during KSHV replication.
Collapse
Affiliation(s)
- Chunhong Di
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, China.,School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Guoxia Zheng
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yunheng Zhang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Enyu Tong
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yanli Ren
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yu Hong
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yang Song
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Rong Chen
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Xiaohua Tan
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Lei Yang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
10
|
Rastgar Rezaei Y, Zarezadeh R, Nikanfar S, Oghbaei H, Nazdikbin N, Bahrami-Asl Z, Zarghami N, Ahmadi Y, Fattahi A, Nouri M, Dittrich R. microRNAs in the pathogenesis of non-obstructive azoospermia: the underlying mechanisms and therapeutic potentials. Syst Biol Reprod Med 2021; 67:337-353. [PMID: 34355990 DOI: 10.1080/19396368.2021.1951890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
miRNAs are involved in different biological processes, including proliferation, differentiation, and apoptosis. Interestingly, 38% of the X chromosome-linked miRNAs are testis-specific and have crucial roles in regulating the renewal and cell cycle of spermatogonial stem cells. Previous studies demonstrated that abnormal expression of spermatogenesis-related miRNAs could lead to nonobstructive azoospermia (NOA). Moreover, differential miRNAs expression in seminal plasma of NOA patients has been reported compared to normozoospermic men. However, the role of miRNAs in NOA pathogenesis and the underlying mechanisms have not been comprehensively studied. Therefore, the aim of this review is to mechanistically describe the role of miRNAs in the pathogenesis of NOA and discuss the possibility of using the miRNAs as therapeutic targets.Abbreviations: AMO: anti-miRNA antisense oligonucleotide; AZF: azoospermia factor region; CDK: cyclin-dependent kinase; DAZ: deleted in azoospermia; ESCs: embryonic stem cells; FSH: follicle-stimulating hormone; ICSI: intracytoplasmic sperm injection; JAK/STAT: Janus kinase/signal transducers and activators of transcription; miRNA: micro-RNA; MLH1: Human mutL homolog l; NF-κB: Nuclear factor-kappa B; NOA: nonobstructive azoospermia; OA: obstructive azoospermia; PGCs: primordial germ cells; PI3K/AKT: Phosphatidylinositol 3-kinase/protein kinase B; Rb: retinoblastoma tumor suppressor; ROS: Reactive Oxygen Species; SCOS: Sertoli cell-only syndrome; SIRT: sirtuin; SNPs: single nucleotide polymorphisms; SSCs: spermatogonial stem cells; TESE: testicular sperm extraction; TGF-β: transforming growth factor-beta.
Collapse
Affiliation(s)
- Yeganeh Rastgar Rezaei
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Nikanfar
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Oghbaei
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Zahra Bahrami-Asl
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Ahmadi
- Department of Urology, Sina Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
Cao G, Gao Z, Jiang Y, Chu M. Lin28 gene and mammalian puberty. Mol Reprod Dev 2020; 87:525-533. [PMID: 32363678 DOI: 10.1002/mrd.23347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/21/2020] [Indexed: 11/10/2022]
Abstract
Lin28a and Lin28b, homologs of the Caenorhabditis elegans Lin28 gene, play important roles in cell pluripotency, reprogramming, and tumorigenicity. Recently, genome-wide association and transgenic studies showed that Lin28a and/or Lin28b gene were involved in the onset of mammalian puberty, the stage representing the attainment of reproduction capacity; however, the detailed mechanism of these genes in mammalian puberty remains largely unknown. The present paper reviews the research progress on the roles of Lin28a/b genes in the onset of mammalian puberty by analyzing the results coming from gene expression patterns, mutations, and transgenic studies, and put forward possible pathways for further studies on their roles in animal reproduction.
Collapse
Affiliation(s)
- Guiling Cao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China.,College of Agriculture, Liaocheng University, Liaocheng, China
| | - Zeyang Gao
- College of Agriculture, Liaocheng University, Liaocheng, China
| | - Yunliang Jiang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Mingxing Chu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
12
|
Ma L, Yu H, Wang X, Li D, Zhang Y, Pei X, Duan Z, Ma M. The effects of maternal exposure to BPA during pregnancy on the male reproductive system and the testicular microRNA expression profile. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:17290-17302. [PMID: 32157532 DOI: 10.1007/s11356-020-08156-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 02/19/2020] [Indexed: 06/10/2023]
Abstract
The effect of prenatal bisphenol A (BPA) exposure is increasingly concerned. We investigated the effect of maternal BPA exposure during pregnancy on male offspring and its potential mechanism. Thirty pregnant Sprague Dawley (SD) rats were randomly divided into exposed and control groups. At PND56, the number of sperm, luteinizing hormone, and testosterone in the BPA-exposed group decreased, and testicular tissue structure was damaged in offsprings. At GD20, the miRNA profile in the testis of male offspring was examined and the expression levels of 28 deregulated miRNAs were validated by qRT-PCR. We found that miR-361-5p, miR-203a-3p, and miR-19b-2-5p had significantly different expression levels in the testis. These results suggest that maternal exposure to BPA can lead to differential changes in progeny miRNAs, which will provide direction for future in-depth mechanisms of reproductive injury.
Collapse
Affiliation(s)
- Lin Ma
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China
| | - Haiyang Yu
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China
| | - Xu Wang
- Department of Environmental Engineering and Bioengineering, Shenyang Institute of Science and Technology, Shenyang, 110167, Liaoning Province, People's Republic of China
| | - Dan Li
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China
| | - Yumin Zhang
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China
| | - Xiucong Pei
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China
| | - Zhiwen Duan
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China
| | - Mingyue Ma
- Department of Toxicology, School of Public Heath, Shenyang Medical College, Shenyang, 110034, Liaoning Province, People's Republic of China.
| |
Collapse
|
13
|
Qi Y, Zheng G, Di C, Zhang J, Wang X, Hong Y, Song Y, Chen R, Yang Y, Yan Y, Xu L, Tan X, Yang L. Latency-associated nuclear antigen inhibits lytic replication of Kaposi's sarcoma-associated herpesvirus by regulating let-7a/RBPJ signaling. Virology 2019; 531:69-78. [PMID: 30856484 DOI: 10.1016/j.virol.2019.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 10/27/2022]
Abstract
Latency-associated nuclear antigen (LANA) is the key factor in the establishment and maintenance of latency of Kaposi's sarcoma-associated herpesvirus (KSHV). A cellular protein, recombination signal binding protein for immunoglobulin kappa J region (RBPJ), is essential for the lytic reactivation of KSHV. However, whether RBPJ expression is regulated by KSHV is not clear. Here, we show that LANA upregulates let-7a and its primary transcripts in parallel with its reduction of RBPJ expression. An increase in notch intracellular domain (NICD) and the downregulation of NF-κB and LIN28B contribute to the upregulation of let-7a by LANA. Let-7a represses RBPJ expression by directly binding the 3' untranslated region of RBPJ. Let-7a overexpression or RBPJ knockdown led to a dose- and time-dependent inhibition of lytic reactivation of KSHV. Collectively, these findings support a model wherein LANA inhibits the lytic replication of KSHV by regulating let-7a/RBPJ signaling.
Collapse
Affiliation(s)
- Yan Qi
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Guoxia Zheng
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Chunhong Di
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Jinxia Zhang
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaobo Wang
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Yu Hong
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Yang Song
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Rong Chen
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Yi Yang
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Yutao Yan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Liangwen Xu
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaohua Tan
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China.
| | - Lei Yang
- School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
14
|
Leinonen JT, Chen YC, Tukiainen T, Panula P, Widén E. Transient modification of lin28b expression - Permanent effects on zebrafish growth. Mol Cell Endocrinol 2019; 479:61-70. [PMID: 30196135 DOI: 10.1016/j.mce.2018.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 08/09/2018] [Accepted: 09/01/2018] [Indexed: 11/24/2022]
Abstract
Recent genome-wide association studies and mouse models have identified LIN28B as a gene affecting several pubertal timing-related traits and vertebrate growth. However, the exact biological mechanisms underlying the associations remain unknown. We have explored the mechanisms linking LIN28B with growth regulation by combining human gene expression data with functional models. Specifically, we show that 1) pubertal timing-associated genetic variation correlates with LIN28B expression in the pituitary and hypothalamus, 2) downregulating lin28b in zebrafish embryos associates with aberrant development of kiss2-neurons, and 3) increasing lin28b expression transiently by synthetic mRNA injections during embryogenesis results in sustained enhancement of zebrafish growth. Unexpectedly, the mRNA injections resulted in advanced sexual maturation of female fish, suggesting that lin28b may influence pubertal timing through multiple developmental mechanisms. Overall, these results provide novel insight into LIN28B function in vertebrate growth regulation, emphasizing the importance of the gene and related genetic pathways for embryonic and juvenile development.
Collapse
Affiliation(s)
- Jaakko T Leinonen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, P.O. Box 20 (Tukholmankatu 8), Helsinki, 00014, Finland
| | - Yu-Chia Chen
- Department of Anatomy and Neuroscience Center, University of Helsinki, P.O. Box 63, (Haartmaninkatu 8), Helsinki, 00014, Finland
| | - Taru Tukiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, P.O. Box 20 (Tukholmankatu 8), Helsinki, 00014, Finland
| | - Pertti Panula
- Department of Anatomy and Neuroscience Center, University of Helsinki, P.O. Box 63, (Haartmaninkatu 8), Helsinki, 00014, Finland
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, P.O. Box 20 (Tukholmankatu 8), Helsinki, 00014, Finland.
| |
Collapse
|
15
|
Xing F, Zhang C, Kong Z. Cloning and expression of lin-28 homolog B gene in the onset of puberty in Duolang sheep. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2018; 32:23-30. [PMID: 30381750 PMCID: PMC6325404 DOI: 10.5713/ajas.18.0276] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 09/14/2018] [Indexed: 11/27/2022]
Abstract
Objective Recent studies have demonstrated that lin-28 homolog B (LIN28B)/miRNA let-7 (let-7) plays a role in the regulation of pubertal onset in mammals. However, the role of LIN28B/let-7 in the onset of ovine puberty remains unknown. We cloned the Duolang sheep Lin28B cDNA sequence, detected the expression change of LIN28B, let-7a and let-7g in hypothalamus, pituitary and ovary tissues at three different pubertal stages. Methods The reverse transcriptase polymerase chain reaction (RT-PCR) was used to clone the cDNA sequence of LIN28B gene from Duolang sheep and the bioinformatics methods were applied to analyze the amino acid sequence of LIN28B protein. The mRNA expression levels of the LIN28B gene at different pubertal stages were examined by real time RT-PCR. Results LIN28B cDNA of Duolang sheep was cloned, and two transcripts were obtained. The amino acid sequence of transcript 1 shares 99.60%, 98.78%, and 94.80% identity with those of goat, wild yak and pig, respectively. Strong LIN28B mRNA expression was detected in the hypothalamus, pituitary, ovary, oviduct and uterus, while moderate expression was found in the liver, kidney, spleen and heart, weak expression was observed in the heart. No expression was found in the lungs. Quantitative real-time PCR (QPCR) and western-blot analysis revealed that the LIN28B was highly expressed in the hypothalamus and ovary at prepuberty stages, and this expression significantly decreased from the prepuberty to puberty stages (p<0.05). Markedly increased levels of mRNA expression were detected in the pituitary from prepuberty to puberty (p<0.05) and then significantly decreased from puberty to postpuberty (p<0.05). The expression levels of let-7a and let-7g showed no significant changes among different pubertal stages (p>0.05). Conclusion These results provided a foundation for determining the functions of LIN28B/let-7 and their role in the onset of sheep puberty.
Collapse
Affiliation(s)
- Feng Xing
- College of Animal Science, Tarim University, Alar, XinJiang 843300, China.,Key laboratory of Tarim, Animal Husbandry Science and Technology, XinJiang Production & Construction Corps, Alar, Xinjiang 843300, China
| | - Chaoyang Zhang
- College of Animal Science, Tarim University, Alar, XinJiang 843300, China.,Key laboratory of Tarim, Animal Husbandry Science and Technology, XinJiang Production & Construction Corps, Alar, Xinjiang 843300, China
| | - Zhengquan Kong
- College of Animal Science, Tarim University, Alar, XinJiang 843300, China.,Key laboratory of Tarim, Animal Husbandry Science and Technology, XinJiang Production & Construction Corps, Alar, Xinjiang 843300, China
| |
Collapse
|
16
|
Precocious Puberty and the Lin28/Let7 Pathway: The Therapeutic Effect of the Nourishing "Yin" and Purging "Fire" Traditional Chinese Medicine Mixture in a Rat Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:4868045. [PMID: 30046338 PMCID: PMC6038664 DOI: 10.1155/2018/4868045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022]
Abstract
The present study aims to investigate the effects of the nourishing “Yin” and purging “Fire” Traditional Chinese Medicine (TCM) herb mixture on precocious puberty and TCM may act through hypothalamic Lin28/let7 pathway expression in the precocious puberty model rats. Meanwhile, to confirm the relationship between Lin28/let7 pathway and puberty by overexpression Lin28a, in the first part of this study, female rats were randomly allocated into untreated controls, the precocious puberty (PP) model group, the PP control group, and the PP + TCM group. Rats on postnatal day 5 were injected danazol to establish the PP model. From days 15 to 35, the rats in the TCM group were given the TCM twice daily. Vaginal opening, sex-related hormones, and body and reproductive organ weights were measured, and the expressions of hypothalamic Lin28a and Lin28b mRNA and let7a and let7b miRNA were detected. In addition, in the second part, the effects of overexpression of Lin28a on the vaginal opening time were evaluated. In the two parts of the study, we found that, at the onset of puberty, a decrease in ovary weight, an increase in the serum levels of luteinizing hormone and progesterone, and increased expression levels of hypothalamic Lin28b mRNA were observed in the PP + TCM group compared to the PP model group. The vaginal opening time was significantly delayed upon overexpression of Lin28a. Above all, the mechanism by which the TCM treats precocious puberty is thus likely to be associated with inhibition of the hypothalamic Lin28/let7 signaling pathway and our findings provide in-depth insight into the relationship between the overexpression of Lin28a gene in the hypothalamus and the onset of puberty.
Collapse
|
17
|
Reproductive role of miRNA in the hypothalamic-pituitary axis. Mol Cell Neurosci 2018; 88:130-137. [DOI: 10.1016/j.mcn.2018.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/19/2017] [Accepted: 01/21/2018] [Indexed: 12/21/2022] Open
|
18
|
Avendaño MS, Vazquez MJ, Tena-Sempere M. Disentangling puberty: novel neuroendocrine pathways and mechanisms for the control of mammalian puberty. Hum Reprod Update 2018; 23:737-763. [PMID: 28961976 DOI: 10.1093/humupd/dmx025] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Puberty is a complex developmental event, controlled by sophisticated regulatory networks that integrate peripheral and internal cues and impinge at the brain centers driving the reproductive axis. The tempo of puberty is genetically determined but is also sensitive to numerous modifiers, from metabolic and sex steroid signals to environmental factors. Recent epidemiological evidence suggests that the onset of puberty is advancing in humans, through as yet unknown mechanisms. In fact, while much knowledge has been gleaned recently on the mechanisms responsible for the control of mammalian puberty, fundamental questions regarding the intimate molecular and neuroendocrine pathways responsible for the precise timing of puberty and its deviations remain unsolved. OBJECTIVE AND RATIONALE By combining data from suitable model species and humans, we aim to provide a comprehensive summary of our current understanding of the neuroendocrine mechanisms governing puberty, with particular focus on its central regulatory pathways, underlying molecular basis and mechanisms for metabolic control. SEARCH METHODS A comprehensive MEDLINE search of articles published mostly from 2003 to 2017 has been carried out. Data from cellular and animal models (including our own results) as well as clinical studies focusing on the pathophysiology of puberty in mammals were considered and cross-referenced with terms related with central neuroendocrine mechanisms, metabolic control and epigenetic/miRNA regulation. OUTCOMES Studies conducted during the last decade have revealed the essential role of novel central neuroendocrine pathways in the control of puberty, with a prominent role of kisspeptins in the precise regulation of the pubertal activation of GnRH neurosecretory activity. In addition, different transmitters, including neurokinin-B (NKB) and, possibly, melanocortins, have been shown to interplay with kisspeptins in tuning puberty onset. Alike, recent studies have documented the role of epigenetic mechanisms, involving mainly modulation of repressors that target kisspeptins and NKB pathways, as well as microRNAs and the related binding protein, Lin28B, in the central control of puberty. These novel pathways provide the molecular and neuroendocrine basis for the modulation of puberty by different endogenous and environmental cues, including nutritional and metabolic factors, such as leptin, ghrelin and insulin, which are known to play an important role in pubertal timing. WIDER IMPLICATIONS Despite recent advancements, our understanding of the basis of mammalian puberty remains incomplete. Complete elucidation of the novel neuropeptidergic and molecular mechanisms summarized in this review will not only expand our knowledge of the intimate mechanisms responsible for puberty onset in humans, but might also provide new tools and targets for better prevention and management of pubertal deviations in the clinical setting.
Collapse
Affiliation(s)
- M S Avendaño
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n. 14004 Córdoba, Spain.,Hospital Universitario Reina Sofia, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain
| | - M J Vazquez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n. 14004 Córdoba, Spain.,Hospital Universitario Reina Sofia, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain
| | - M Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n. 14004 Córdoba, Spain.,Hospital Universitario Reina Sofia, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| |
Collapse
|
19
|
Johannessen C, Moi L, Kiselev Y, Pedersen MI, Dalen SM, Braaten T, Busund LT. Expression and function of the miR-143/145 cluster in vitro and in vivo in human breast cancer. PLoS One 2017; 12:e0186658. [PMID: 29073169 PMCID: PMC5657998 DOI: 10.1371/journal.pone.0186658] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/01/2017] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that function as post-transcriptional regulators of gene expression and are dysregulated in cancer. Studies of miRNAs to explore their potential as diagnostic and prognostic markers are of great scientific interest. Here, we investigate the functional properties and expression of the miR-143/145 cluster in breast cancer (BC) in vitro and in vivo. The ER positive MCF7, the HER2 positive SK-BR-3, and the triple negative cell line MDA-MB-231 were used to assess cell proliferation and cell invasion. Expression of miRNA in 108 breast cancers in the Norwegian Women and Cancer Study and 44 benign tissue controls were analyzed by microarray and validated by RT-PCR. Further, in situ hybridization (ISH) was used to study the cellular and subcellular distribution of the miRNAs. In vitro, miR-143 promoted proliferation of MCF7 and MDA-MB-231 cells, whereas miR-145 and the cotransfection of both miRNAs inhibited proliferation in all three cell lines. The cells’ invasive capacity was reduced after transfection and cotransfection of the miRNAs. In line with the tumor suppressive functions in vitro, the expression of miR-143 and miR-145 was lower in malignant compared to benign breast tissue, and lower in the more aggressive tumors with higher tumor grade, loss of ER and the basal-like phenotype. ISH revealed miR-143 to be cytoplasmatic and predominantly expressed in luminal cells in benign tissue, whilst miR-145 was nuclear and with strong staining in myoepithelial cells. Both miRNAs were present in malignant epithelial cells and stromal fibroblasts in BC. This study demonstrates that miR-143 and -145 have functional properties and expression patterns typical for tumor suppressors, but the function is influenced by cellular factors such as cell type and miRNA cotransfection. Further, the nuclear functions of miR-145 should be explored for a more complete understanding of the complexity of miRNA regulation and function in BC.
Collapse
Affiliation(s)
- Charles Johannessen
- Department of Medical Biology, UiT—The Arctic University of Norway, Tromsø, Norway
- * E-mail:
| | - Line Moi
- Department of Medical Biology, UiT—The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Yury Kiselev
- Department of Life Sciences and Health, Oslo and Akershus University College of Applied Sciences, Oslo, Norway
| | - Mona Irene Pedersen
- Department of Clinical Medicine, UiT—The Arctic University of Norway, Tromsø, Norway
| | - Stig Manfred Dalen
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Tonje Braaten
- Department of Community Medicine, UiT—The Arctic University of Norway, Tromsø, Norway
| | - Lill-Tove Busund
- Department of Medical Biology, UiT—The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
20
|
Skaftnesmo KO, Edvardsen RB, Furmanek T, Crespo D, Andersson E, Kleppe L, Taranger GL, Bogerd J, Schulz RW, Wargelius A. Integrative testis transcriptome analysis reveals differentially expressed miRNAs and their mRNA targets during early puberty in Atlantic salmon. BMC Genomics 2017; 18:801. [PMID: 29047327 PMCID: PMC5648517 DOI: 10.1186/s12864-017-4205-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Our understanding of the molecular mechanisms implementing pubertal maturation of the testis in vertebrates is incomplete. This topic is relevant in Atlantic salmon aquaculture, since precocious male puberty negatively impacts animal welfare and growth. We hypothesize that certain miRNAs modulate mRNAs relevant for the initiation of puberty. To explore which miRNAs regulate mRNAs during initiation of puberty in salmon, we performed an integrated transcriptome analysis (miRNA and mRNA-seq) of salmon testis at three stages of development: an immature, long-term quiescent stage, a prepubertal stage just before, and a pubertal stage just after the onset of single cell proliferation activity in the testis. RESULTS Differentially expressed miRNAs clustered into 5 distinct expression profiles related to the immature, prepubertal and pubertal salmon testis. Potential mRNA targets of these miRNAs were predicted with miRmap and filtered for mRNAs displaying negatively correlated expression patterns. In summary, this analysis revealed miRNAs previously known to be regulated in immature vertebrate testis (miR-101, miR-137, miR-92b, miR-18a, miR-20a), but also miRNAs first reported here as regulated in the testis (miR-new289, miR-30c, miR-724, miR-26b, miR-new271, miR-217, miR-216a, miR-135a, miR-new194 and the novel predicted n268). By KEGG enrichment analysis, progesterone signaling and cell cycle pathway genes were found regulated by these differentially expressed miRNAs. During the transition into puberty we found differential expression of miRNAs previously associated (let7a/b/c), or newly associated (miR-15c, miR-2184, miR-145 and the novel predicted n7a and b) with this stage. KEGG enrichment analysis revealed that mRNAs of the Wnt, Hedgehog and Apelin signaling pathways were potential regulated targets during the transition into puberty. Likewise, several regulated miRNAs in the pubertal stage had earlier been associated (miR-20a, miR-25, miR-181a, miR-202, let7c/d/a, miR-125b, miR-222a/b, miR-190a) or have now been found connected (miR-2188, miR-144, miR-731, miR-8157 and the novel n2) to the initiation of puberty. CONCLUSIONS This study has - for the first time - linked testis maturation to specific miRNAs and their inversely correlated expressed targets in Atlantic salmon. The study indicates a broad functional conservation of already known miRNAs and associated pathways involved in the transition into puberty in vertebrates. The analysis also reveals miRNAs not previously associated with testis tissue or its maturation, which calls for further functional studies in the testis.
Collapse
Affiliation(s)
- K O Skaftnesmo
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway.
| | - R B Edvardsen
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - T Furmanek
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - D Crespo
- Reproductive Biology group, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - E Andersson
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - L Kleppe
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - G L Taranger
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - J Bogerd
- Reproductive Biology group, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - R W Schulz
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway.,Reproductive Biology group, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - A Wargelius
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| |
Collapse
|
21
|
Rao YS, Pak TR. microRNAs and the adolescent brain: Filling the knowledge gap. Neurosci Biobehav Rev 2016; 70:313-322. [PMID: 27328787 PMCID: PMC5074866 DOI: 10.1016/j.neubiorev.2016.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/09/2016] [Accepted: 06/11/2016] [Indexed: 12/14/2022]
Abstract
Over two decades ago the discovery of microRNAs (miRNA) broadened our understanding of the diverse molecular pathways mediating post-transcriptional control over gene expression. These small non-coding RNAs dynamically fluctuate, temporally and spatially, throughout the lifespan of all organisms. The fundamental role that miRNAs have in shaping embryonic neurodevelopment provides strong evidence that adolescent brain remodeling could be rooted in the changing miRNA landscape of the cell. Few studies have directly measured miRNA gene expression changes in the brain across pubertal development, and even less is known about the functional impact of those miRNAs on the maturational processes that occur in the developing adolescent brain. This review summarizes miRNA biogenesis and function in the brain in the context of normal (i.e. not diseased) physiology. These landmark studies can guide predictions about the role of miRNAs in facilitating maturation of the adolescent brain. However, there are clear indicators that adolescence/puberty is a unique life stage, suggesting miRNA function during adolescence is distinct from those in any other previously described system.
Collapse
Affiliation(s)
- Yathindar S Rao
- Loyola University Chicago, Stritch School of Medicine, Department of Cell and Molecular Physiology, United States
| | - Toni R Pak
- Loyola University Chicago, Stritch School of Medicine, Department of Cell and Molecular Physiology, United States.
| |
Collapse
|
22
|
Faunes F, Larraín J. Conservation in the involvement of heterochronic genes and hormones during developmental transitions. Dev Biol 2016; 416:3-17. [DOI: 10.1016/j.ydbio.2016.06.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/03/2016] [Accepted: 06/09/2016] [Indexed: 01/26/2023]
|
23
|
Corre C, Shinoda G, Zhu H, Cousminer DL, Crossman C, Bellissimo C, Goldenberg A, Daley GQ, Palmert MR. Sex-specific regulation of weight and puberty by the Lin28/let-7 axis. J Endocrinol 2016; 228:179-91. [PMID: 26698568 PMCID: PMC4772724 DOI: 10.1530/joe-15-0360] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
Growth and pubertal timing differ in boys and girls. Variants in/near LIN28B associate with age at menarche (AAM) in genome-wide association studies and some AAM-related variants associate with growth in a sex-specific manner. Sex-specific growth patterns in response to Lin28b perturbation have been detected in mice, and overexpression of Lin28a has been shown to alter pubertal timing in female mice. To investigate further how Lin28a and Lin28b affect growth and puberty in both males and females, we evaluated Lin28b loss-of-function (LOF) mice and Lin28a gain-of-function (GOF) mice. Because both Lin28a and Lin28b can act via the conserved microRNA let-7, we also examined let-7 GOF mice. As reported previously, Lin28b LOF led to lighter body weights only in male mice while Lin28a GOF yielded heavier mice of both sexes. Let-7 GOF mice weighed less than controls, and males were more affected than females. Timing of puberty was assessed by vaginal opening (VO) and preputial separation (PS). Male Lin28b LOF and male let-7 GOF, but not female, mice displayed alteration of pubertal timing, with later PS than controls. In contrast, both male and female Lin28a GOF mice displayed late onset of puberty. Together, these data point toward a complex system of regulation by Lin28a, Lin28b, and let-7, in which Lin28b and let-7 can impact both puberty and growth in a sex-specific manner, raising the possibility that this pathway may contribute to differential regulation of male and female growth and puberty in humans.
Collapse
Affiliation(s)
- Christina Corre
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Gen Shinoda
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Hao Zhu
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Diana L Cousminer
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Christine Crossman
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Christian Bellissimo
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Anna Goldenberg
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - George Q Daley
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Mark R Palmert
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|