156 results on '"Lee B. Smith"'
Search Results
2. LetsTalkShots: personalized vaccine risk communication
- Author
-
Daniel A. Salmon, Matthew Z. Dudley, Janesse Brewer, Jana Shaw, Holly B. Schuh, Tina M. Proveaux, Amelia M. Jamison, Amanda Forr, Michelle Goryn, Robert F. Breiman, Walter A. Orenstein, Lee-Sien Kao, Robina Josiah Willock, Michelle Cantu, Tori Decea, Robin Mowson, Kate Tsubata, Lucie Marisa Bucci, Jaqueline Lawler, James D. Watkins, Jamie W. Moore, James H. Fugett, Adriele Fugal, Yazmine Tovar, Marie Gay, Aleen M. Cary, Iulia Vann, Lee B. Smith, Lilly Kan, Magda Mankel, Sumayya Beekun, Victoria Smith, Stephanie D. Adams, Steven A. Harvey, and Peter Z. Orton
- Subjects
vaccine hesitancy ,communication ,COVID-19 ,vaccines ,tailored application ,Public aspects of medicine ,RA1-1270 - Abstract
IntroductionVaccine hesitancy is a global health threat undermining control of many vaccine-preventable diseases. Patient-level education has largely been ineffective in reducing vaccine concerns and increasing vaccine uptake. We built and evaluated a personalized vaccine risk communication website called LetsTalkShots in English, Spanish and French (Canadian) for vaccines across the lifespan. LetsTalkShots tailors lived experiences, credible sources and informational animations to disseminate the right message from the right messenger to the right person, applying a broad range of behavioral theories.MethodsWe used mixed-methods research to test our animation and some aspects of credible sources and personal narratives. We conducted 67 discussion groups (n = 325 persons), stratified by race/ethnicity (African American, Hispanic, and White people) and population (e.g., parents, pregnant women, adolescents, younger adults, and older adults). Using a large Ipsos survey among English-speaking respondents (n = 2,272), we tested animations aligned with vaccine concerns and specific to population (e.g., parents of children, parents of adolescents, younger adults, older adults).ResultsDiscussion groups provided robust feedback specific to each animation as well as areas for improvements across animations. Most respondents indicated that the information presented was interesting (85.5%), clear (96.0%), helpful (87.0%), and trustworthy (82.2%).DiscussionTailored vaccine risk communication can assist decision makers as they consider vaccination for themselves, their families, and their communities. LetsTalkShots presents a model for personalized communication in other areas of medicine and public health.
- Published
- 2023
- Full Text
- View/download PDF
3. Sertoli cell-enriched proteins in mouse and human testicular interstitial fluid
- Author
-
Liza O’Donnell, Laura F. Dagley, Michael Curley, Annalucia Darbey, Peter J. O’Shaughnessy, Thorsten Diemer, Adrian Pilatz, Daniela Fietz, Peter G. Stanton, Lee B. Smith, and Diane Rebourcet
- Subjects
Medicine ,Science - Abstract
Sertoli cells support the development of sperm and the function of various somatic cells in the interstitium between the tubules. Sertoli cells regulate the function of the testicular vasculature and the development and function of the Leydig cells that produce testosterone for fertility and virility. However, the Sertoli cell-derived factors that regulate these cells are largely unknown. To define potential mechanisms by which Sertoli cells could support testicular somatic cell function, we aimed to identify Sertoli cell-enriched proteins in the testicular interstitial fluid (TIF) between the tubules. We previously resolved the proteome of TIF in mice and humans and have shown it to be a rich source of seminiferous tubule-derived proteins. In the current study, we designed bioinformatic strategies to interrogate relevant proteomic and genomic datasets to identify Sertoli cell-enriched proteins in mouse and human TIF. We analysed proteins in mouse TIF that were significantly reduced after one week of acute Sertoli cell ablation in vivo and validated which of these are likely to arise primarily from Sertoli cells based on relevant mouse testis RNASeq datasets. We used a different, but complementary, approach to identify Sertoli cell-enriched proteins in human TIF, taking advantage of high-quality human testis genomic, proteomic and immunohistochemical datasets. We identified a total of 47 and 40 Sertoli cell-enriched proteins in mouse and human TIF, respectively, including 15 proteins that are conserved in both species. Proteins with potential roles in angiogenesis, the regulation of Leydig cells or steroidogenesis, and immune cell regulation were identified. The data suggests that some of these proteins are secreted, but that Sertoli cells also deposit specific proteins into TIF via the release of extracellular vesicles. In conclusion, we have identified novel Sertoli cell-enriched proteins in TIF that are candidates for regulating somatic cell-cell communication and testis function.
- Published
- 2023
4. A role for steroid 5 alpha-reductase 1 in vascular remodeling during endometrial decidualization
- Author
-
Isaac W. Shaw, Phoebe M. Kirkwood, Diane Rebourcet, Fiona L. Cousins, Rebecca J. Ainslie, Dawn E. W. Livingstone, Lee B. Smith, Philippa T.K. Saunders, and Douglas A. Gibson
- Subjects
decidualization ,angiogenesis ,dihydrotestosterone ,5 alpha-reductase ,androgen ,intracrinology ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Decidualization is the hormone-dependent process of endometrial remodeling that is essential for fertility and reproductive health. It is characterized by dynamic changes in the endometrial stromal compartment including differentiation of fibroblasts, immune cell trafficking and vascular remodeling. Deficits in decidualization are implicated in disorders of pregnancy such as implantation failure, intra-uterine growth restriction, and pre-eclampsia. Androgens are key regulators of decidualization that promote optimal differentiation of stromal fibroblasts and activation of downstream signaling pathways required for endometrial remodeling. We have shown that androgen biosynthesis, via 5α-reductase-dependent production of dihydrotestosterone, is required for optimal decidualization of human stromal fibroblasts in vitro, but whether this is required for decidualization in vivo has not been tested. In the current study we used steroid 5α-reductase type 1 (SRD5A1) deficient mice (Srd5a1-/- mice) and a validated model of induced decidualization to investigate the role of SRD5A1 and intracrine androgen signaling in endometrial decidualization. We measured decidualization response (weight/proportion), transcriptomic changes, and morphological and functional parameters of vascular development. These investigations revealed a striking effect of 5α-reductase deficiency on the decidualization response. Furthermore, vessel permeability and transcriptional regulation of angiogenesis signaling pathways, particularly those that involved vascular endothelial growth factor (VEGF), were disrupted in the absence of 5α-reductase. In Srd5a1-/- mice, injection of dihydrotestosterone co-incident with decidualization restored decidualization responses, vessel permeability, and expression of angiogenesis genes to wild type levels. Androgen availability declines with age which may contribute to age-related risk of pregnancy disorders. These findings show that intracrine androgen signaling is required for optimal decidualization in vivo and confirm a major role for androgens in the development of the vasculature during decidualization through regulation of the VEGF pathway. These findings highlight new opportunities for improving age-related deficits in fertility and pregnancy health by targeting androgen-dependent signaling in the endometrium.
- Published
- 2022
- Full Text
- View/download PDF
5. New Insights into Testosterone Biosynthesis: Novel Observations from HSD17B3 Deficient Mice
- Author
-
Ben M. Lawrence, Liza O’Donnell, Lee B. Smith, and Diane Rebourcet
- Subjects
androgens ,testosterone ,HSD17B3 ,enzymes ,canonical pathway ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Androgens such as testosterone and dihydrotestosterone (DHT) are essential for male sexual development, masculinisation, and fertility. Testosterone is produced via the canonical androgen production pathway and is essential for normal masculinisation and testis function. Disruption to androgen production can result in disorders of sexual development (DSD). In the canonical pathway, 17β-hydroxysteroid dehydrogenase type 3 (HSD17B3) is viewed as a critical enzyme in the production of testosterone, performing the final conversion required. HSD17B3 deficiency in humans is associated with DSD due to low testosterone concentration during development. Individuals with HSD17B3 mutations have poorly masculinised external genitalia that can appear as ambiguous or female, whilst having internal Wolffian structures and testes. Recent studies in mice deficient in HSD17B3 have made the surprising finding that testosterone production is maintained, male mice are masculinised and remain fertile, suggesting differences between mice and human testosterone production exist. We discuss the phenotypic differences observed and the possible other pathways and enzymes that could be contributing to testosterone production and male development. The identification of alternative testosterone synthesising enzymes could inform the development of novel therapies to endogenously regulate testosterone production in individuals with testosterone deficiency.
- Published
- 2022
- Full Text
- View/download PDF
6. A Novel Model Using AAV9-Cre to Knockout Adult Leydig Cell Gene Expression Reveals a Physiological Role of Glucocorticoid Receptor Signalling in Leydig Cell Function
- Author
-
Anne-Louise Gannon, Annalucia L. Darbey, Grace Chensee, Ben M. Lawrence, Liza O’Donnell, Joanna Kelso, Natalie Reed, Shanmathi Parameswaran, Sarah Smith, Lee B. Smith, and Diane Rebourcet
- Subjects
glucocorticoid receptor ,androgens ,leydig cells ,AAV9 ,steroidogenesis ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Glucocorticoids are steroids involved in key physiological processes such as development, metabolism, inflammatory and stress responses and are mostly used exogenously as medications to treat various inflammation-based conditions. They act via the glucocorticoid receptor (GR) expressed in most cells. Exogenous glucocorticoids can negatively impact the function of the Leydig cells in the testis, leading to decreased androgen production. However, endogenous glucocorticoids are produced by the adrenal and within the testis, but whether their action on GR in Leydig cells regulates steroidogenesis is unknown. This study aimed to define the role of endogenous GR signalling in adult Leydig cells. We developed and compared two models; an inducible Cre transgene driven by expression of the Cyp17a1 steroidogenic gene (Cyp17-iCre) that depletes GR during development and a viral vector-driven Cre (AAV9-Cre) to deplete GR in adulthood. The delivery of AAV9-Cre ablated GR in adult mouse Leydig cells depleted Leydig cell GR more efficiently than the Cyp17-iCre model. Importantly, adult depletion of GR in Leydig cells caused reduced expression of luteinising hormone receptor (Lhcgr) and of steroidogenic enzymes required for normal androgen production. These findings reveal that Leydig cell GR signalling plays a physiological role in the testis and highlight that a normal balance of glucocorticoid activity in the testis is important for steroidogenesis.
- Published
- 2022
- Full Text
- View/download PDF
7. Specific Transcriptomic Signatures and Dual Regulation of Steroidogenesis Between Fetal and Adult Mouse Leydig Cells
- Author
-
Pauline Sararols, Isabelle Stévant, Yasmine Neirijnck, Diane Rebourcet, Annalucia Darbey, Michael K. Curley, Françoise Kühne, Emmanouil Dermitzakis, Lee B. Smith, and Serge Nef
- Subjects
fetal Leydig cell ,adult Leydig cell ,androgen ,testis ,RNA sequencing ,single cell RNA sequencing ,Biology (General) ,QH301-705.5 - Abstract
Leydig cells (LC) are the main testicular androgen-producing cells. In eutherian mammals, two types of LCs emerge successively during testicular development, fetal Leydig cells (FLCs) and adult Leydig cells (ALCs). Both display significant differences in androgen production and regulation. Using bulk RNA sequencing, we compared the transcriptomes of both LC populations to characterize their specific transcriptional and functional features. Despite similar transcriptomic profiles, a quarter of the genes show significant variations in expression between FLCs and ALCs. Non-transcriptional events, such as alternative splicing was also observed, including a high rate of intron retention in FLCs compared to ALCs. The use of single-cell RNA sequencing data also allowed the identification of nine FLC-specific genes and 50 ALC-specific genes. Expression of the corticotropin-releasing hormone 1 (Crhr1) receptor and the ACTH receptor melanocortin type 2 receptor (Mc2r) specifically in FLCs suggests a dual regulation of steroidogenesis. The androstenedione synthesis by FLCs is stimulated by luteinizing hormone (LH), corticotrophin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH) whereas the testosterone synthesis by ALCs is dependent exclusively on LH. Overall, our study provides a useful database to explore LC development and functions.
- Published
- 2021
- Full Text
- View/download PDF
8. Androgen Receptor Is Dispensable for X-Zone Regression in the Female Adrenal but Regulates Post-Partum Corticosterone Levels and Protects Cortex Integrity
- Author
-
Anne-Louise Gannon, Laura O’Hara, Ian J. Mason, Anne Jørgensen, Hanne Frederiksen, Michael Curley, Laura Milne, Sarah Smith, Rod T. Mitchell, and Lee B. Smith
- Subjects
adrenal cortex ,X-zone ,androgen receptor ,spindle cell ,hyperplasia ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Adrenal androgens are fundamental mediators of ovarian folliculogenesis, embryonic implantation, and breast development. Although adrenal androgen function in target tissues are well characterized, there is little research covering the role of androgen-signaling within the adrenal itself. Adrenal glands express AR which is essential for the regression of the X-zone in male mice. Female mice also undergo X-zone regression during their first pregnancy, however whether this is also controlled by AR signaling is unknown. To understand the role of the androgen receptor (AR) in the female adrenal, we utilized a Cyp11a1-Cre to specifically ablate AR from the mouse adrenal cortex. Results show that AR-signaling is dispensable for adrenal gland development in females, and for X-zone regression during pregnancy, but is required to suppress elevation of corticosterone levels post-partum. Additionally, following disruption to adrenal AR, aberrant spindle cell development is observed in young adult females. These results demonstrate sexually dimorphic regulation of the adrenal X-zone by AR and point to dysfunctional adrenal androgen signaling as a possible mechanism in the early development of adrenal spindle cell hyperplasia.
- Published
- 2021
- Full Text
- View/download PDF
9. Modelling steroidogenesis: a framework model to support hypothesis generation and testing across endocrine studies
- Author
-
Laura O’Hara, Peter J. O’Shaughnessy, Tom C. Freeman, and Lee B. Smith
- Subjects
Steroidogenesis ,Model ,Diagram ,Medicine ,Biology (General) ,QH301-705.5 ,Science (General) ,Q1-390 - Abstract
Abstract Objective Steroid hormones are responsible for the control of a wide range of physiological processes such as development, growth, reproduction, metabolism, and aging. Because of the variety of enzymes, substrates and products that take part in steroidogenesis and the compartmentalisation of its constituent reactions, it is a complex process to visualise and document. One of the goals of systems biology is to quantitatively describe the behaviour of complex biological systems that involve the interaction of many components. This can be done by representing these interactions visually in a pathway model and then optionally constructing a mathematical model of the interactions. Results We have used the modified Edinburgh Pathway Notation to construct a framework diagram describing human steroidogenic pathways, which will be of use to endocrinologists. To demonstrate further utility, we show how such models can be parameterised with empirical data within the software Graphia Professional, to recapitulate specific examples of steroid hormone production, and also to mimic gene knockout. These framework models support in silico hypothesis generation and testing with utility across endocrine endpoints, with significant potential to reduce costs, time and animal numbers, whilst informing the design of planned studies.
- Published
- 2018
- Full Text
- View/download PDF
10. Deciphering Cell Lineage Specification during Male Sex Determination with Single-Cell RNA Sequencing
- Author
-
Isabelle Stévant, Yasmine Neirijnck, Christelle Borel, Jessica Escoffier, Lee B. Smith, Stylianos E. Antonarakis, Emmanouil T. Dermitzakis, and Serge Nef
- Subjects
single-cell RNA-seq ,sex determination ,testis ,Sertoli cell ,fetal Leydig cell ,progenitors ,Biology (General) ,QH301-705.5 - Abstract
Summary: The gonad is a unique biological system for studying cell-fate decisions. However, major questions remain regarding the identity of somatic progenitor cells and the transcriptional events driving cell differentiation. Using time-series single-cell RNA sequencing on XY mouse gonads during sex determination, we identified a single population of somatic progenitor cells prior to sex determination. A subset of these progenitors differentiates into Sertoli cells, a process characterized by a highly dynamic genetic program consisting of sequential waves of gene expression. Another subset of multipotent cells maintains their progenitor state but undergoes significant transcriptional changes restricting their competence toward a steroidogenic fate required for the differentiation of fetal Leydig cells. Our findings confirm the presence of a unique multipotent progenitor population in the gonadal primordium that gives rise to both supporting and interstitial lineages. These also provide the most granular analysis of the transcriptional events occurring during testicular cell-fate commitment.
- Published
- 2018
- Full Text
- View/download PDF
11. Low-dose tamoxifen treatment in juvenile males has long-term adverse effects on the reproductive system: implications for inducible transgenics
- Author
-
Saloni H. Patel, Laura O’Hara, Nina Atanassova, Sarah E. Smith, Michael K. Curley, Diane Rebourcet, Annalucia L. Darbey, Anne-Louise Gannon, Richard M. Sharpe, and Lee B. Smith
- Subjects
Medicine ,Science - Abstract
Abstract The tamoxifen-inducible Cre system is a popular transgenic method for controlling the induction of recombination by Cre at a specific time and in a specific cell type. However, tamoxifen is not an inert inducer of recombination, but an established endocrine disruptor with mixed agonist/antagonist activity acting via endogenous estrogen receptors. Such potentially confounding effects should be controlled for, but >40% of publications that have used tamoxifen to generate conditional knockouts have not reported even the minimum appropriate controls. To highlight the importance of this issue, the present study investigated the long-term impacts of different doses of a single systemic tamoxifen injection on the testis and the wider endocrine system. We found that a single dose of tamoxifen less than 10% of the mean dose used for recombination induction, caused adverse effects to the testis and to the reproductive endocrine system that persisted long-term. These data raise significant concerns about the widespread use of tamoxifen induction of recombination, and highlight the importance of including appropriate controls in all pathophysiological studies using this means of induction.
- Published
- 2017
- Full Text
- View/download PDF
12. Sertoli cells as key drivers of testis function
- Author
-
Diane Rebourcet, Lee B. Smith, and Liza O'Donnell
- Subjects
Male ,endocrine system ,Sertoli Cells ,Leydig cell ,urogenital system ,medicine.drug_class ,Somatic cell ,Leydig Cells ,Cell Biology ,Biology ,Androgen ,Sertoli cell ,Sperm ,Rats ,Cell biology ,medicine.anatomical_structure ,Immune privilege ,Testis ,medicine ,Animals ,Humans ,Spermatogenesis ,Germ cell ,Developmental Biology - Abstract
Sertoli cells are the orchestrators of spermatogenesis; they support fetal germ cell commitment to the male pathway and are essential for germ cell development, from maintenance of the spermatogonial stem cell niche and spermatogonial populations, through meiosis and spermiogeneis and to the final release of mature spermatids during spermiation. However, Sertoli cells are also emerging as key regulators of other testis somatic cells, including supporting peritubular myoid cell development in the pre-pubertal testis and supporting the function of the testicular vasculature and in contributing to testicular immune privilege. Sertoli cells also have a major role in regulating androgen production within the testis, by specifying interstitial cells to a steroidogenic fate, contributing to androgen production in the fetal testis, and supporting fetal and adult Leydig cell development and function. Here, we provide an overview of the specific roles for Sertoli cells in the testis and highlight how these cells are key drivers of testicular sperm output, and of adult testis size and optimal function of other testicular somatic cells, including the steroidogenic Leydig cells.
- Published
- 2022
13. Hyperprolactinemia in a male pituitary androgen receptor knockout mouse is associated with female‐like lactotroph development
- Author
-
Lee B. Smith, Helen C. Christian, Paul Le Tissier, and Laura O’Hara
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Lactotrophs ,medicine.drug_class ,Urology ,Endocrinology, Diabetes and Metabolism ,Biology ,Prolactin cell ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Anterior pituitary ,Internal medicine ,medicine ,Animals ,Testosterone ,Mice, Knockout ,030219 obstetrics & reproductive medicine ,Estrogens ,Androgen ,Prolactin ,Hyperprolactinemia ,Androgen receptor ,medicine.anatomical_structure ,Reproductive Medicine ,Receptors, Androgen ,Hypothalamus ,Estrogen ,Pituitary Gland ,hormones, hormone substitutes, and hormone antagonists - Abstract
Background Circulating prolactin concentration in rodents and humans is sexually dimorphic. Estrogens are a well-characterised stimulator of prolactin release. Circulating prolactin fluctuates throughout the menstrual/estrous cycle of females in response to estrogen levels, but remains continually low in males. We have previously identified androgens as an inhibitor of prolactin release through characterisation of males of a mouse line with a conditional pituitary androgen receptor knockout (PARKO) which have an increase in circulating prolactin, but unchanged lactotroph number. Objectives In the present study we aimed to specify the cell type that androgens act on to repress prolactin release. Materials and methods PARKO, lactotroph-specific, Pit1 lineage-specific and neural-specific conditional androgen receptor knockout male mice were investigated using prolactin ELISA, pituitary electron microscopy, immunohistochemistry and qRT-PCR. Results Lactotroph-specific, Pit1 lineage-specific and neural-specific conditional AR knockouts did not duplicate the high circulating prolactin seen in the PARKO line. Using electron microscopy to examine ultrastructure we showed that pituitary androgen receptor knockout male mice develop lactotrophs that resemble those seen in female mice. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males. When expression of selected estrogen-regulated anterior pituitary genes were examined there were no differences in expression level between controls and knockouts. Discussion The cell type that androgens act on to repress prolactin release is not the lactotroph, cells in the Pit1-lineage, or the dopaminergic neurons in the hypothalamus. PARKO males develop a female-specific lactotroph ultrastructure that this is likely to contribute to the increase in circulating prolactin. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males, which suggests that removal of both circulating estrogens and androgens reduces the stimulation of pituitary prolactin release. Conclusion Further investigation is needed into prolactin regulation by changes in androgen-estrogen balance, which is involved sexual dimorphism of development and diseases including hyperprolactinemia. This article is protected by copyright. All rights reserved.
- Published
- 2021
14. A comparison of in vivo viral targeting systems identifies adeno‐associated virus serotype 9 (AAV9) as an effective vector for genetic manipulation of Leydig cells in adult mice
- Author
-
Nathan Jeffery, Karen R. Kilcoyne, Annalucia Darbey, Lee B. Smith, Diane Rebourcet, Michael Curley, Pamela Brown, Natalie L Reed, Laura Milne, and Cornelia Roesl
- Subjects
Male ,Cell type ,viruses ,Urology ,Endocrinology, Diabetes and Metabolism ,Transgene ,Genetic Vectors ,medicine.disease_cause ,Adenoviridae ,Viral vector ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,medicine ,Animals ,Vector (molecular biology) ,Adeno-associated virus ,030219 obstetrics & reproductive medicine ,biology ,Leydig cell ,Lentivirus ,Gene Transfer Techniques ,Leydig Cells ,Dependovirus ,biology.organism_classification ,Cell biology ,medicine.anatomical_structure ,Reproductive Medicine ,biology.protein ,Neuraminidase - Abstract
Background Despite the increasing popularity of deliverable transgenics, a robust and fully validated method for targeting Leydig cells, capable of delivering long-term transgene expression, is yet to be defined. Objectives We compared three viral vector systems in terms of their cell targeting specificity, longevity of gene expression and impact on targeted cell types when delivered to the interstitial compartment of the mouse testis. Materials & methods We delivered lentiviral, adenoviral and adeno-associated (AAV) viral particles to the interstitial compartment of adult mouse testis. Immunolocalization and stereology were performed to characterize ability of vectors to target and deliver transgenes to Leydig cells. Results Viral vectors utilized in this study were found to specifically target Leydig cells when delivered interstitially. Transgene expression in lentiviral-targeted Leydig cells was detected for 7 days post-injection before Leydig cells underwent apoptosis. Adenoviral-delivered transgene expression was detected for 10 days post-injection with no evidence of targeted cell apoptosis. We found serotype differences in AAV injected testis with AAV serotype 9 targeting a significant proportion of Leydig cells. Targeting efficiency increased to an average of 59.63% (and a maximum of 80%) of Leydig cells with the addition of neuraminidase during injection. In AAV injected testis sections, transgene expression was detectable for up to 50 days post-injection. Discussion & conclusion Lentivirus, Adenovirus and Adeno-Associated virus delivery to the testis resulted in key variances in targeting efficiency of Leydig cells and in longevity of transgene expression, but identified AAV9 + Neuraminidase as an efficient vector system for transgene delivery and long-term expression. Simple viral delivery procedures and the commercial availability of viral vectors suggests AAV9 + Neuraminidase will be of significant utility to researchers investigating the genetics underpinning Leydig cell function and holds promise to inform the development of novel therapeutics for the treatment of male reproductive disorders.
- Published
- 2020
15. Physical, cognitive, and mental health impacts of COVID-19 after hospitalisation (PHOSP-COVID): a UK multicentre, prospective cohort study
- Author
-
Helen McShane, A Alamoudi, D Parekh, G Burns, R. Gisli Jenkins, Marco Sereno, R Djukanovic, Onn Min Kon, N I Lone, David M. Evans, L Daines, N A Hanley, Z. Omar, William Greenhalf, Nicola Williams, K Storton, Asaf David, T Wallis, E Pacpaco, H McCauley, L. O'Brien, K Hainey, P. Novotny, C Tong, L Ingram, S Gurram, C Avram, C Coleman, Edward T. Bullmore, Richard G. Brown, R Aul, K A Tripp, D. Cristiano, A Michael, Michael C Steiner, Padmasayee Papineni, A Howell, Gail Carson, Peter J. M. Openshaw, Simon Heller, G Madzamba, K Paradowski, S Singh, K Bramham, Teresa Light, David Price, V Shaw, A Yousuf, T Dong, T Hiwot, G Simons, Philip L. Molyneaux, A Ashworth, Ashley C. Brown, N Magee, A L Tan, R A Evans, Mark Toshner, Robert Sykes, W Saxon, S Finney, A Mohamed, P Cairns, Christos P Kotanidis, J D Chalmers, O Adeyemi, L Knibbs, A J Moss, S L Rowland-Jones, O M Kon, L P Ho, A Martineau, B Zhao, M G Crooks, J Meiring, Ewen M Harrison, Louise V. Wain, S Wright, E Robertson, David A. Lomas, H Lamlum, David E. Newby, P Chowdhury, K Mangion, Toby Hillman, E Turner, H McAllister-Williams, S West, J McGinness, B Whittam, T. Gorsuch, K Dempsey, L Mcgarvey, K Poinasamy, K Shevket, Emma Baldry, M Buch, N French, Olivia C. Leavy, Stephen C. J. Parker, H Newell, Louise M. Howard, O. Zongo, P Beirne, C Sharpe, N Mills, C David, M Bayley, Carmine M. Pariante, P Haldar, Z Kausar, A Dipper, I Hall, P McArdle, G Ogg, Rachael A. Evans, A.J. Buttress, M Pareek, Paul E Pfeffer, Denise Anderson, James D. Chalmers, P Kar, Caroline J. Jolley, S Plein, Nigel J. Brunskill, C Oliver, John R. Hurst, Clive Ballard, F Barrett, D Baguley, Nick P. Talbot, N Chaudhuri, A Young, Jonathan P. Busby, H Dobson, K Holmes, Liam G Heaney, Ruth E Barker, Anthony N. Price, David J. Stensel, L Brear, Louise Sigfrid, Marcia Soares, Patrice Carter, J R Hurst, John R. Geddes, Donghyung Lee, L Watson, J M Lord, H Parfrey, N Odell, J Glossop, K. Liyanage, Bryan Williams, S Neubauer, O Elneima, David R Baldwin, G Mallison, C Francis, A Te, D Foote, F Woodhead, A De Soyza, A Atkins, M Stern, A Morley, E Bright, N Basu, Simon E. Brill, D Southern, D Forton, L G Heaney, B Raman, Malcolm G Semple, M Mariveles, Charalambos Antoniades, Nawar Diar Bakerly, Swapna Mandal, Aroon D. Hingorani, E K Sage, Ania Korszun, A Hosseini, Louise Allan, M Toshner, Fergus V. Gleeson, Cherie Armour, J Quigley, S Drain, Thomas Kabir, M Havinden-Williams, Ben G. Marshall, S Patale, C Bourne, L Wright, Rachel L. Batterham, S Jones, S Linford, Salman Siddiqui, C Laing, A Horsley, S Greenwood, A Lingford-Hughes, S. Jose, Stefan Neubauer, S L Dobson, M Rahman, Alex D. McMahon, S Young, A Frankel, Joe Dennis, Claire M. Nolan, J Fuld, J Mayet, Nayia Petousi, Brij Patel, A Fairman, F Speranza, A Bularga, Colin Berry, Charlotte L. Edwardson, A Lloyd, H Jones, N Mairs, H Assefa-Kebede, L Gilmour, D Jones, Siobhan Kelly, I Cruz, Tim Rees, A Haggar, R. Wolf-Roberts, R Flockton, R Dowling, Geraldine Landers, C. Price, P Neill, John B. Cole, A L Key, Elaine Hardy, P Kitterick, Elodie Murali, Carly Welch, P Crisp, Rachel C. Chambers, L Carr, P C Calder, A McQueen, S Defres, A Dewar, F Adeyemi, Avan Aihie Sayer, D W Connell, M Halling-Brown, Neil J. Greening, M Andrews, Linda MacLiver, Kevin A. Davies, E Wade, Elizabeth M. Tunnicliffe, H Jarvis, Kathryn M. Abel, N Hart, A J Yousuf, Nicholas Easom, Alexander Richards, Lee B. Smith, P Dulawan, Janet T Scott, Amisha Singapuri, E Sapey, G Willis, P M George, S Bain, H. Tench, S S Kon, N Window, M J Rowland, A. J. Shah, B Card, A Knighton, P Chowienczyk, Luke Daines, Cathie Sudlow, Joseph Jacob, J Rossdale, S Paddick, Ifan Jones, A Storrie, Sonia Johnson, Huzaifa Adamali, Gail Davies, R G Jenkins, J Murira, Kamlesh Khunti, W Y James, Ajay M. Shah, A B Docherty, Donna J. Menzies, R Morriss, K Piper Hanley, James J Furniss, C Overton, P Mansoori, Phil Harrison, P Greenhaff, A Humphries, H. McGuinness, Gerome Breen, Hayley Hardwick, Davies Adeloye, P Pfeffer, H Lota, Daniel G. Wootton, William Monteiro, A Holbourn, R Hamil, Y Ellis, Traolach S. Brugha, A Alli, D Wraith, Jennifer K Quint, H Atkins, I Peralta, David C. Thomas, A Bolger, J Rodgers, S Portukhay, David Wilson, Michael Sharpe, Steven Kerr, T Plekhanova, J Lewis, S. Quaid, O Olaosebikan, L Lim, K Roy, A Checkley, A Newton Cox, A Dougherty, Bill Deakin, R Pius, A Hoare, N. Dormand, T Craig, Dhruv Parekh, Betty Raman, K E Lewis, Christopher E. Brightling, L G Spencer, Z Suleiman, E R Chilvers, Keith M. Channon, A Saratzis, R Lenagh, N Diar Bakerly, I Macharia, G Kaltsakas, L Morrison, M Ralser, K Fallon, C J Tee, JM Watson, J Nunag, R Gregory, J E Pearl, C Wright, K Regan, D Johnston, P Hogarth, Najib M. Rahman, G P McCann, Julie Evans, N Easom, Joseph Hughes, J Skeemer, H Baxendale, E Hufton, B Elliott, L V Wain, Ardythe L. Morrow, Meenal Patel, S Glover, C Xie, M Harvie, Alan Hughes, David B. Thomas, N Choudhury, Mark J. Tobin, Elizabeta B. Mukaetova-Ladinska, Richard W. Francis, J L Heeney, Shyam Madathil, Ellen Guthrie, S Yasmin, H Turton, M Marks, I Koychev, Melanie J. Davies, John P Greenwood, Daniel Peckham, E Lee, Iain B. McInnes, K Hadley, Charlotte Summers, J Chen, A Prickett, Timothy R Nicholson, K Lewis, A Cross, Jamie Brown, G Ross, H Wheeler, Manu Sharma, Igor Rudan, A Routen, M J Noonan, J Wild, K Jiwa, B. Welsh, Jonathan Pimm, J Kwan, A Lucey, C Favager, K Brindle, Nazir I Lone, Naveed Sattar, C Christie, James E. Mitchell, M Wilkins, C Coupland, T Thornton, Christian P Subbe, Alex Horsley, J Blaikely, G F Toingson, S Walsh, A Lea, Jennifer A. Smith, Margot W. Parkes, M Dixon, Luke Howard, N Majeed, A Hayday, Jack A. Sargeant, Michael Pavlides, K Leitch, J. Pendlebury, Andrew Donaldson, T Peto, Thomas A Jackson, N Rahman, M Gibbons, J Phipps, S Logan, D Wilkinson, J Breeze, D Holgate, R Osbourne, M Hoare, M Malim, Ryan S Thwaites, Stephen R Knight, W Ibrahim, J Rowland, Andrew M. Taylor, B Al-Sheklly, R. Loosley, S Megson, C Summersgill, Z Coburn, R Evans, I Wilson, B Pathmanathan, Jeremy George, A Angyal, S Betts, A Deans, C E Brightling, S Kerr, N Selby, L Price, A Ramos, S N Diwanji, P Kurupati, J S Brown, K Scott, A Sheikh, Krisnah Poinasamy, R Ugwuoke, Teresa Thompson, K Chong-James, Gerry P McCann, John R. Petrie, R Hughes, E. Watson, K McIvor, Trudie Chalder, Melissa Heightman, B Gooptu, H Evans, Thomas Yates, R Ahmed, Nicholas Hart, R Allen, W Schwaeble, J Simpson, Sara Clohisey, Janet M. Lord, R Bell, R Baggott, Clare J Taylor, Keir Lewis, Lynda Connor, F Thaivalappil, Kathryn J Saunders, Lynsey S. Hall, Richard Kevin Stone, Aliki Thomas, L Turtle, H Tedd, L Matthews, J Bambrough, S Stanel, M J McMahon, L Chetham, Enya Daynes, R Hurst, Angela Cook, M Aljaroof, Ling-Pei Ho, Paul Moss, H Arnold, S Fairbairn, Anthony J. Rostron, L Garner, Kyle Harrington, Douglas Grieve, B Connolly, Khalida Ismail, Craig Johnson, E. Russell, T Hussell, S Kon, Claudia Langenberg, E Wall, A Rowland, Miriam Harvey, N Powell, Catherine Pennington, N Armstrong, J C Porter, A Ient, Matthew Hotopf, R Parvin, M Richardson, I Smith, L Lightstone, J. Dasgin, Lynne Armstrong, A Charalambou, J R Geddes, C. Clark, E Gourlay, A Botkai, G Choudhury, J Bonnington, Matthew A. Brown, Paul Dark, S Thackray-Nocera, J Woods, E Stringer, R Free, Aarti Shikotra, J Jacob, P Clift, W Man, Sally J Singh, B King, Nikki Gautam, A Zawia, K McCafferty, L Milligan, S Whittaker, A Elmer, H Chinoy, H Welch, J Haworth, A Shikotra, Matthew J. Rowland, A Singapuri, M McNarry, F. Davies, F Khan, T Mcnally, Alfred A.R. Thompson, A McArdle, V Brown, Helen L. Fisher, M Spears, Peter Jezzard, Morag Henderson, D Thickett, U Munawar, M Broome, Graeme Jones, M. Gummadi, S Marciniak, L Poll, E Calvelo, J Hawkes, D Saralaya, S Walder, Omer Elneima, E M Harrison, C E Bolton, S J Singh, Khalid Shah, S Diver, M Willicombe, M Ainsworth, H Nassa, O C Leavy, D C Thomas, R Upthegrove, C Singh, C Echevarria, Sebastian Edwards, N Lewis-Burke, C Bloomfield, D L Sykes, J Parmar, Sam M. Janes, Simon Wessely, Shaney L Barratt, Judith Clarke, S McAdoo, G MacGowan, Hamish McAuley, L O Wajero, C Dobson, David J. Burn, Daniel Lasserson, Gill Arbane, Matthew Richardson, D McAulay, Rhian M. Touyz, Miles D. Witham, E Major, J Whitney, C J Jolley, Michael Beadsworth, N Goodman, S Walmsley, Daniel F. McAuley, Kath Chapman, Paul Cullinan, Margaret Jones, K P Yip, Nilesh J. Samani, M Bourne, Jeremy S. Brown, A Bloss, Alison M. Lawrie, Timothy Felton, L Bishop, T Sass, Oliver Polgar, M Bakali, N Hawkings, T Chalder, Mujtaba Husain, B Jayaraman, Hannah Bayes, Vicky Kamwa, B Hargadon, Y Peng, C Jolley, D Matila, Clare E. Mackay, J Worsley, R Dharmagunawardena, R Samuel, L Fabbri, R Russell, K Bhui, David W. Clark, S Heller, Anne Dell, J Nyaboko, N Huneke, Michael Marks, L Hesselden, A Greenhalgh, L Broad, M Bakau, Susan P. Walker, Marlies Ostermann, Smitaa Patel, E Fraser, R I Evans, V Whitehead, S Ahmad, C King, B Young, David T Arnold, Paul Klenerman, S Dunn, H McAuley, D Faluyi, B Holroyd-Hind, H Qureshi, E Bradley, Brendan G Cooper, P Shah, L Houchen, Shelley Fletcher, Todd Evans, Andrew Smith, Jill Walsh, Amanda F. Elliott, V Harris, L Holdsworth, A Ford, R Saunders, K Vellore, Jonathan Finch, A McGovern, D Nicoll, A Briggs, J Oxton, G A Davies, Milton Ashworth, T I de Silva, V Lewis, James Stockley, S Byrne, Alison G. Harvey, M Sereno, Marc Lipman, S Terry, A Moss, L. McMorrow, Nick A Maskell, Annemarie B Docherty, R Sabit, J Kenneth Baillie, Jennifer M. Short, Louise Stadon, Aziz Sheikh, S A Williams-Howard, Atul Gupta, D Altmann, J Cavanagh, S Francis, E. Perkins, E McIvor, P Atkin, Julie Williams, D Sutherland, J Rangeley, Derek Bell, J Valabhji, J K Baillie, Isobel D. Stewart, P McCourt, P Rivera-Ortega, N J Greening, Anthony De Soyza, M Dalton, Group, PHOSP-COVID Collaborative, Apollo - University of Cambridge Repository, Baguley, David, National Institute for Health Research, UKRI MRC COVID-19 Rapid Response Call, and UK Research and Innovation
- Subjects
Adult ,Male ,Pulmonary and Respiratory Medicine ,Pediatrics ,medicine.medical_specialty ,Health Status ,medicine.medical_treatment ,MEDLINE ,Comorbidity ,Disease ,Logistic regression ,PHOSP-COVID Collaborative Group ,1117 Public Health and Health Services ,Cognition ,SDG 3 - Good Health and Well-being ,medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Aged ,Mechanical ventilation ,United Kingdom/epidemiology ,business.industry ,COVID-19 ,1103 Clinical Sciences ,Articles ,Middle Aged ,Mental health ,United Kingdom ,Middle age ,Hospitalization ,Mental Health ,Acute Disease ,COVID-19/complications ,Female ,business ,1199 Other Medical and Health Sciences ,Follow-Up Studies - Abstract
Background The impact of COVID-19 on physical and mental health and employment after hospitalisation with acute disease is not well understood. The aim of this study was to determine the effects of COVID-19-related hospitalisation on health and employment, to identify factors associated with recovery, and to describe recovery phenotypes. Methods The Post-hospitalisation COVID-19 study (PHOSP-COVID) is a multicentre, long-term follow-up study of adults (aged ≥18 years) discharged from hospital in the UK with a clinical diagnosis of COVID-19, involving an assessment between 2 and 7 months after discharge, including detailed recording of symptoms, and physiological and biochemical testing. Multivariable logistic regression was done for the primary outcome of patient-perceived recovery, with age, sex, ethnicity, body-mass index, comorbidities, and severity of acute illness as covariates. A post-hoc cluster analysis of outcomes for breathlessness, fatigue, mental health, cognitive impairment, and physical performance was done using the clustering large applications k-medoids approach. The study is registered on the ISRCTN Registry (ISRCTN10980107). Findings We report findings for 1077 patients discharged from hospital between March 5 and Nov 30, 2020, who underwent assessment at a median of 5·9 months (IQR 4·9–6·5) after discharge. Participants had a mean age of 58 years (SD 13); 384 (36%) were female, 710 (69%) were of white ethnicity, 288 (27%) had received mechanical ventilation, and 540 (50%) had at least two comorbidities. At follow-up, only 239 (29%) of 830 participants felt fully recovered, 158 (20%) of 806 had a new disability (assessed by the Washington Group Short Set on Functioning), and 124 (19%) of 641 experienced a health-related change in occupation. Factors associated with not recovering were female sex, middle age (40–59 years), two or more comorbidities, and more severe acute illness. The magnitude of the persistent health burden was substantial but only weakly associated with the severity of acute illness. Four clusters were identified with different severities of mental and physical health impairment (n=767): very severe (131 patients, 17%), severe (159, 21%), moderate along with cognitive impairment (127, 17%), and mild (350, 46%). Of the outcomes used in the cluster analysis, all were closely related except for cognitive impairment. Three (3%) of 113 patients in the very severe cluster, nine (7%) of 129 in the severe cluster, 36 (36%) of 99 in the moderate cluster, and 114 (43%) of 267 in the mild cluster reported feeling fully recovered. Persistently elevated serum C-reactive protein was positively associated with cluster severity. Interpretation We identified factors related to not recovering after hospital admission with COVID-19 at 6 months after discharge (eg, female sex, middle age, two or more comorbidities, and more acute severe illness), and four different recovery phenotypes. The severity of physical and mental health impairments were closely related, whereas cognitive health impairments were independent. In clinical care, a proactive approach is needed across the acute severity spectrum, with interdisciplinary working, wide access to COVID-19 holistic clinical services, and the potential to stratify care. Funding UK Research and Innovation and National Institute for Health Research.
- Published
- 2021
16. Leukemia inhibitory factor-receptor signalling negatively regulates gonadotrophin-stimulated testosterone production in mouse Leydig Cells
- Author
-
Michael Curley, Annalucia Darbey, Liza O'Donnell, Karen R. Kilcoyne, Kirsten Wilson, Will Mungall, Diane Rebourcet, Jingtao Guo, Rod T. Mitchell, and Lee B. Smith
- Subjects
Male ,Mice ,Endocrinology ,Receptors, OSM-LIF ,Testis ,Animals ,Leydig Cells ,Testosterone ,Molecular Biology ,Biochemistry ,Leukemia Inhibitory Factor - Abstract
Testicular Leydig cells (LCs) are the principal source of circulating testosterone in males. LC steroidogenesis maintains sexual function, fertility and general health, and is influenced by various paracrine factors. The leukemia inhibitory factor receptor (LIFR) is expressed in the testis and activated by different ligands, including leukemia inhibitory factor (LIF), produced by peritubular myoid cells. LIF can modulate LC testosterone production in vitro under certain circumstances, but the role of consolidated signalling through LIFR in adult LC function in vivo has not been established. We used a conditional Lifr allele in combination with adenoviral vectors expressing Cre-recombinase to generate an acute model of LC Lifr-KO in the adult mouse testis, and showed that LC Lifr is not required for short term LC survival or basal steroidogenesis. However, LIFR-signalling negatively regulates steroidogenic enzyme expression and maximal gonadotrophin-stimulated testosterone biosynthesis, expanding our understanding of the intricate regulation of LC steroidogenic function.
- Published
- 2021
17. Novel Role for p110β PI 3-Kinase in Male Fertility through Regulation of Androgen Receptor Activity in Sertoli Cells.
- Author
-
Julie Guillermet-Guibert, Lee B Smith, Guillaume Halet, Maria A Whitehead, Wayne Pearce, Diane Rebourcet, Kelly León, Pascale Crépieux, Gemma Nock, Maria Strömstedt, Malin Enerback, Claude Chelala, Mariona Graupera, John Carroll, Sabina Cosulich, Philippa T K Saunders, Ilpo Huhtaniemi, and Bart Vanhaesebroeck
- Subjects
Genetics ,QH426-470 - Abstract
The organismal roles of the ubiquitously expressed class I PI3K isoform p110β remain largely unknown. Using a new kinase-dead knockin mouse model that mimics constitutive pharmacological inactivation of p110β, we document that full inactivation of p110β leads to embryonic lethality in a substantial fraction of mice. Interestingly, the homozygous p110β kinase-dead mice that survive into adulthood (maximum ~26% on a mixed genetic background) have no apparent phenotypes, other than subfertility in females and complete infertility in males. Systemic inhibition of p110β results in a highly specific blockade in the maturation of spermatogonia to spermatocytes. p110β was previously suggested to signal downstream of the c-kit tyrosine kinase receptor in germ cells to regulate their proliferation and survival. We now report that p110β also plays a germ cell-extrinsic role in the Sertoli cells (SCs) that support the developing sperm, with p110β inactivation dampening expression of the SC-specific Androgen Receptor (AR) target gene Rhox5, a homeobox gene critical for spermatogenesis. All extragonadal androgen-dependent functions remain unaffected by global p110β inactivation. In line with a crucial role for p110β in SCs, selective inactivation of p110β in these cells results in male infertility. Our study is the first documentation of the involvement of a signalling enzyme, PI3K, in the regulation of AR activity during spermatogenesis. This developmental pathway may become active in prostate cancer where p110β and AR have previously been reported to functionally interact.
- Published
- 2015
- Full Text
- View/download PDF
18. The expanded roles of Sertoli cells: lessons from Sertoli cell ablation models
- Author
-
Peter J. O'Shaughnessy, Lee B. Smith, and Diane Rebourcet
- Subjects
0301 basic medicine ,endocrine system ,Cell type ,urogenital system ,Endocrinology, Diabetes and Metabolism ,Transgene ,030209 endocrinology & metabolism ,Biology ,Sertoli cell ,Cell biology ,Transplantation ,03 medical and health sciences ,Adult life ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cell culture ,Isolation techniques ,medicine - Abstract
Since Enrico Sertoli's first depiction of Sertoli cells in the 19th century, the role of Sertoli cells as a hub for both testis biology and as a potential therapeutic tool, continues to grow. The function of Sertoli cells in the orchestration of testis differentiation and their role in nurturing germ cells in adult life has been well characterised in recent years. The combination of cell culture, isolation techniques and the development of a plethora of transgenic models has put an emphasis on the inter-cellular dialogue and regulation between Sertoli and the other testicular cell types. More recently, the use of a Sertoli cell ablation model has identified a number of unexpected functions carried out by the Sertoli cells during testis development and in the adult. The accumulation of data has not only unlocked a new paradigm in reproductive function and regulation but also guides new strategies in transplantation approaches. In this review, we summarise the recently identified, expanded roles of Sertoli cells and describe their importance in relation to topics such as endocrine disruption, infertility and new tools for therapy.
- Published
- 2019
19. Sperm-specific proteins: new implications for diagnostic development and cancer immunotherapy
- Author
-
Liza, O'Donnell, Lee B, Smith, and Diane, Rebourcet
- Subjects
Male ,Semen ,Neoplasms ,Humans ,Immunotherapy ,Cell Biology ,Seminiferous Tubules ,Spermatozoa - Abstract
Spermatozoa are comprised of many unique proteins not expressed elsewhere. Sperm-specific proteins are first expressed at puberty, after the development of immune tolerance to self-antigens, and have been assumed to remain confined inside the seminiferous tubules, protected from immune cell recognition by various mechanisms of testicular immune privilege. However, new data has shown that sperm-specific proteins are released by the tubules into the surrounding interstitial fluid; from here they can contact immune cells, potentially promote immune tolerance, and enter the circulation. These new findings have clinical implications for diagnostics and therapeutics targeted at a specific class of proteins known as cancer-testis antigens (CTA), the opportunity to identify new communication pathways in the testis, and to discover new ways to monitor testis function.
- Published
- 2022
20. Specific transcriptomic signatures and dual regulation of steroidogenesis between fetal and adult mouse Leydig cells
- Author
-
Lee B. Smith, Diane Rebourcet, Isabelle Stévant, Emmanouil T. Dermitzakis, Annalucia Darbey, Yasmine Neirijnck, Françoise Kühne, Serge Nef, Pauline Sararols, and Michael Curley
- Subjects
0301 basic medicine ,endocrine system ,medicine.drug_class ,Alternative splicing ,Intron ,Biology ,Androgen ,Cell biology ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,ACTH receptor ,Receptor ,Luteinizing hormone ,Testosterone - Abstract
Leydig cells (LC) are the main testicular androgen-producing cells. In eutherian mammals, two types of LCs emerge successively during testicular development, fetal Leydig cells (FLCs) and adult Leydig cells (ALCs). Both display significant differences in androgen production and regulation. Using bulk RNA sequencing, we compared the transcriptomes of both LC populations to characterise their specific transcriptional and functional features. Despite similar transcriptomic profiles, a quarter of the genes show significant variations in expression between FLCs and ALCs. Non-transcriptional events, such as alternative splicing was also observed, including a high rate of intron retention in FLCs compared to ALCs. The use of single-cell RNA sequencing data also allowed the identification of nine FLC-specific genes and 50 ALC-specific genes. Expression of the corticotropin-releasing hormone 1 (Crhr1) receptor and the ACTH receptor melanocortin type 2 receptor (Mc2r) specifically in FLCs suggests a dual regulation of steroidogenesis. The androstenedione synthesis by FLCs is stimulated by luteinizing hormone (LH), CRH and ACTH whereas the testosterone synthesis by ALCs is dependent exclusively on LH. Overall, our study provides a useful database to explore LC development and function.
- Published
- 2021
21. Sperm proteins and cancer-testis antigens are released by the seminiferous tubules in mice and men
- Author
-
Peter G. Stanton, Lee B. Smith, Julien M. D. Legrand, Giuseppe Infusini, Frédéric Chalmel, Diane Rebourcet, Andrew I. Webb, Raouda Sgaier, Laura F. Dagley, Adrian Pilatz, Robin M. Hobbs, Wolfgang Weidner, Liza O'Donnell, Robert I McLachlan, Thorsten Diemer, Daniela Fietz, Peter J. O'Shaughnessy, Hudson Institute of Medical Research [Clayton], University of Newcastle [Australia] (UoN), The Walter and Eliza Hall Institute of Medical Research (WEHI), Justus-Liebig-Universität Gießen (JLU), Institut de recherche en santé, environnement et travail (Irset), Université d'Angers (UA)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-École des Hautes Études en Santé Publique [EHESP] (EHESP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), University of Edinburgh, BB/J015105/1, Biotechnology and Biological Sciences Research Council, Deutsche Forschungsgemeinschaft, Chard-Hutchinson, Xavier, Justus-Liebig-Universität Gießen = Justus Liebig University (JLU), Université d'Angers (UA)-Université de Rennes (UR)-École des Hautes Études en Santé Publique [EHESP] (EHESP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), and University of Newcastle [Callaghan, Australia] (UoN)
- Subjects
0301 basic medicine ,Male ,Proteome ,medicine.medical_treatment ,interstitial fluid ,[SDV]Life Sciences [q-bio] ,Biochemistry ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Neoplasms ,Research Articles ,Seminiferous Tubules ,Sertoli cell ,Spermatozoa ,cancer‐testis antigen ,3. Good health ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Cancer/testis antigens ,biomarker ,Immunotherapy ,Erratum ,cancer-testis antigen ,Biotechnology ,Research Article ,Biology ,testis ,sperm ,Andrology ,03 medical and health sciences ,Immune system ,Antigen ,Antigens, Neoplasm ,Genetics ,medicine ,Animals ,Humans ,Spermatogenesis ,Molecular Biology ,Blood-Testis Barrier ,Infertility, Male ,Sertoli Cells ,Cancer ,Proteins ,Extracellular Fluid ,medicine.disease ,030104 developmental biology ,Cancer biomarkers ,030217 neurology & neurosurgery - Abstract
International audience; Sperm develop from puberty in the seminiferous tubules, inside the blood-testis barrier to prevent their recognition as "non-self" by the immune system, and it is widely assumed that human sperm-specific proteins cannot access the circulatory or immune systems. Sperm-specific proteins aberrantly expressed in cancer, known as cancer-testis antigens (CTAs), are often pursued as cancer biomarkers and therapeutic targets based on the assumption they are neoantigens absent from the circulation in healthy men. Here, we identify a wide range of germ cell-derived and sperm-specific proteins, including multiple CTAs, that are selectively deposited by the Sertoli cells of the adult mouse and human seminiferous tubules into testicular interstitial fluid (TIF) that is "outside" the blood-testis barrier. From TIF, the proteins can access the circulatory- and immune systems. Disruption of spermatogenesis decreases the abundance of these proteins in mouse TIF, and a sperm-specific CTA is significantly decreased in TIF from infertile men, suggesting that exposure of certain CTAs to the immune system could depend on fertility status. The results provide a rationale for the development of blood-based tests useful in the management of male infertility and indicate CTA candidates for cancer immunotherapy and biomarker development that could show sex-specific and male-fertility-related responses.
- Published
- 2021
22. Testosterone regulation on quiescin sulfhydryl oxidase 2 synthesis in the epididymis
- Author
-
Tse-En Wang, Hiroko Tsukamura, Laura O’Hara, Bart M. Gadella, Fuko Matsuda, Pei-Shiue Tsai, Sheng-Hsiang Li, Lee B. Smith, Kei-ichiro Maeda, and Shiori Minabe
- Subjects
0301 basic medicine ,Male ,Embryology ,medicine.drug_class ,Amino Acid Transport System X-AG ,Glutamine ,Glutamic Acid ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Endocrinology ,Human fertilization ,medicine ,Protein biosynthesis ,Animals ,Oxidoreductases Acting on Sulfur Group Donors ,Testosterone ,Epididymis ,Mice, Inbred ICR ,030219 obstetrics & reproductive medicine ,Chemistry ,Glutamate receptor ,Obstetrics and Gynecology ,Cell Biology ,Androgen ,Sperm ,Cell biology ,Sperm Maturation ,030104 developmental biology ,medicine.anatomical_structure ,Reproductive Medicine ,Gene Expression Regulation ,Carrier Proteins - Abstract
The epididymis is an androgen-responsive organ, whose structure and functions are modulated by the coordination between androgen and epididymal cues. Highly regulated molecular interaction within the epididymis is required to support viable sperm development necessary for subsequent fertilization. In the present study, we extended our earlier findings on a promising epididymal protein, quiescin sulfhydryl oxidase 2 (QSOX2), and demonstrated a positive correlation between testosterone and QSOX2 protein synthesis through the use of loss- and restore-of-function animal models. Moreover, based on transcriptomic analyses and 2D culture system, we determined that an additional polarized effect of glutamate is indispensable for the regulatory action of testosterone on QSOX2 synthesis. In conclusion, we propose noncanonical testosterone signaling supports epididymal QSOX2 protein synthesis, providing a novel perspective on the regulation of sperm maturation within the epididymis.
- Published
- 2020
23. Characterisation of a mural cell network in the murine pituitary gland
- Author
-
Laura O’Hara, Lee B. Smith, Nathan Jeffery, Helen C. Christian, and Paul Le Tissier
- Subjects
0301 basic medicine ,Pituitary gland ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Population ,Enteroendocrine cell ,Cell Communication ,Biology ,Stem cell marker ,Mural cell ,Receptor, Platelet-Derived Growth Factor beta ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Paracrine signalling ,Mice ,0302 clinical medicine ,Endocrinology ,Anterior pituitary ,SOX2 ,Pituitary Gland, Anterior ,Internal medicine ,medicine ,Animals ,education ,education.field_of_study ,Endocrine and Autonomic Systems ,SOXB1 Transcription Factors ,Endothelial Cells ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Pituitary Gland ,Endocrine Cells ,Pericytes ,030217 neurology & neurosurgery ,Biomarkers - Abstract
The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRβ) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRβ+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRβ+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.
- Published
- 2020
24. Reconstitution of rat fetal testis during the masculinisation programming window induces focal dysgenesis consistent with testicular dysgenesis syndrome
- Author
-
Joni Macdonald, Ellie Smart, Lee B. Smith, and Rod T. Mitchell
- Subjects
0301 basic medicine ,Male ,Cord ,Reproductive disorders ,lcsh:Medicine ,Reproductive biology ,Biology ,Androgen suppression ,Immunofluorescence ,Gonadal Dysgenesis ,Article ,Andrology ,03 medical and health sciences ,Dysgenesis ,0302 clinical medicine ,Seminal vesicle ,In vivo ,Pregnancy ,Testis ,medicine ,Animals ,Rats, Wistar ,lcsh:Science ,Fetus ,030219 obstetrics & reproductive medicine ,Multidisciplinary ,medicine.diagnostic_test ,lcsh:R ,Sertoli cell ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Q ,Female - Abstract
Focal dysgenesis is a consistent feature of testicular dysgenesis syndrome (TDS) in humans. Rodent studies show that perturbation of androgens (e.g. following phthalate exposure) during a fetal masculinisation programming window (MPW) predisposes to a TDS phenotype. This study aimed to determine whether dissociation and reconstitution of rat fetal testis tissue during the MPW can be used to model and manipulate seminiferous cord development, including induction of focal dysgenesis, as described in TDS. Dissociated fetal rat testes were xenotransplanted subcutaneously into recipient mice for 4 weeks. Transplanted mice were treated with vehicle or di-n-butyl-phthalate (DBP, a plasticising chemical known to induce testicular dysgenesis in vivo in rats). Testosterone production by the transplants was measured in recipient mice and immunofluorescence was performed on the retrieved transplants to identify features consistent with focal testicular dysgenesis. Re-aggregation of rat fetal testis tissue xenotransplants during the MPW results in reconstitution of seminiferous cords. Features of focal testicular dysgenesis were present in re-aggregated testis, including ectopic Sertoli cells and intratubular Leydig cells (ITLCs). DBP exposure of recipient mice reduced androgen-dependent seminal vesicle weight (8.3 vs 26.7 mg; p 2; p > 0.05). We conclude that seminiferous cord reformation during the MPW results in development of focal dysgenesis. The system may be used to separate specific effects (e.g. androgen suppression) of individual chemical exposures from other mechanisms that may be conserved in TDS.
- Published
- 2020
25. Hyperprolactinemia in a male pituitary androgen receptor knockout mouse model is associated with a female-like pattern of lactotroph development
- Author
-
Laura O’Hara, Lee B. Smith, Paul Le Tissier, and Helen C. Christian
- Subjects
endocrine system ,medicine.medical_specialty ,medicine.drug_class ,Biology ,Androgen ,Prolactin ,Prolactin cell ,Androgen receptor ,Endocrinology ,medicine.anatomical_structure ,Anterior pituitary ,Estrogen ,Internal medicine ,Knockout mouse ,medicine ,hormones, hormone substitutes, and hormone antagonists ,Gene knockout - Abstract
Circulating prolactin concentration in rodents and humans is sexually dimorphic. Estrogens are a well-characterised stimulator of prolactin release. Circulating prolactin fluctuates throughout the menstrual/estrous cycle of females in response to estrogen levels, but remains continually low in males. We have previously identified androgens as an inhibitor of prolactin release through characterisation of males of a mouse line with a conditional pituitary androgen receptor knockout (PARKO) which have an increase in circulating prolactin, but unchanged lactotroph number. In the present study we aimed to specify the cell type that androgens act on to repress prolactin release. We examined lactotroph-specific, Pit1 lineage-specific and neural-specific conditional AR knockouts, however they did not duplicate the high circulating prolactin seen in the pituitary androgen receptor knockout line, suggesting that the site of androgen repression of prolactin production was another cell type. Using electron microscopy to examine ultrastructure we showed that pituitary androgen receptor knockout male mice develop lactotrophs that resemble those seen in female mice, and that this is likely to contribute to the increase in circulating prolactin. When castrated, pituitary androgen receptor knockout males have significantly reduced circulating prolactin compared to intact males, which suggests that removal of circulating estrogens as well as androgens reduces the stimulation of pituitary prolactin release. However, when expression of selected estrogen-regulated anterior pituitary genes were examined there were no differences in expression level between controls and knockouts. Further investigation is needed into prolactin regulation by changes in androgen-estrogen balance, which has implications not only in the normal sexual dimorphism of physiology but also in diseases such as hyperprolactinemia.
- Published
- 2020
26. Ectopic FGF23 production induces mineral changes, osteogenic transdifferentiation, and cancer associated microcalcifications
- Author
-
Ida M Boisen, Peter W. Andrews, Peter J. O'Shaughnessy, Birgitte G. Toft, Lee B Smith, Ireen Kooij, Noriko Ide, Anders Juul, Jovanna Kaludjerovic, Martin Blomberg Jensen, Ewa Rajpert-De Meyts, J E Nielsen, Richard Norman, and Beate Lanske
- Subjects
endocrine system ,Chemistry ,Fibroblast growth factor receptor 1 ,Transdifferentiation ,Sertoli cell ,medicine.disease ,Embryonic stem cell ,Cell biology ,Androgen receptor ,Embryonal carcinoma ,medicine.anatomical_structure ,medicine ,Alkaline phosphatase ,Stem cell - Abstract
Testicular microcalcifications consist of hydroxyapatite and their demonstration by ultrasound has been associated with increased risk of testicular germ cell cancer (TGCT). Here, we show that fibroblast growth factor 23 (FGF23), a bone-specific regulator of phosphate homeostasis, is expressed in testicular germ cell neoplasia in situ (GCNIS), embryonal carcinoma (EC), and human embryonic stem cells. FGF23 is not glycosylated in TGCTs and thus rapidly cleaved into a C-terminal fragment that serves as a competitive antagonist for full-length FGF23. High levels of C-terminal FGF23 occupy the receptor formed by Klotho and FGF receptor 1 (FGFR1) in the germ cells facilitating a shift in the expression of phosphate transport proteins from SLC34A2 to SLC34A1 in seminiferous tubules adjacent to GCNIS. Fgf23 knockout mice have a marked epididymal deposition of hydroxyapatite, while the testicular phenotype is milder with spermatogenic arrest and focal germ-cell-specific expression of the bone-like markers runt-related transcription factor 2 (RUNX2) and bone gamma-carboxyglutamic acid-containing protein (BGLAP). In accordance, mice with no functional androgen receptor and lack of circulating gonadotropins develop microcalcifications in 94% of cases and have lower Slc34a2, and higher Slc34a1 and Bglap expression. In accordance, human testicular specimens with microcalcifications also have lower SLC34A2, and focally germ cells express SLC34A1, BGLAP, and RUNX2. Importantly, calcium or phosphate induced osteogenic transdifferentiation of a spermatogonial cell line in vitro demonstrated by induction of alkaline phosphatase activity and deposition of hydroxyapatite, which could be fully rescued by pyrophosphate (PPi). Severe microcalcifications were also found in a mouse model with Sertoli-cell ablation particularly when Sertoli-ablation was conducted prepubertally where the germ cells retain stem cell potential. In conclusion, cancer-related microcalcifications may arise secondary to gonadal mineral alterations, which in combination with impaired Sertoli cell function and reduced PPi due to high alkaline phosphatase activity in GCNIS and TGCTs, facilitates osteogenic transdifferentiation of testicular germ cells and deposition of hydroxyapatite.
- Published
- 2020
27. Ablation of the canonical testosterone production pathway via knockout of the steroidogenic enzyme HSD17B3, reveals a novel mechanism of testicular testosterone production
- Author
-
Annalucia Darbey, Anne Jørgensen, Diane Rebourcet, Lee B. Smith, Michael Curley, Peter J. O'Shaughnessy, Hanne Frederiksen, Rod T. Mitchell, Serge Nef, and Rosa Mackay
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,17-Hydroxysteroid Dehydrogenases ,Mutant ,Biology ,HSD17B12 ,HSD17B3 ,Biochemistry ,Mice ,03 medical and health sciences ,Basal (phylogenetics) ,Leydig cell ,0302 clinical medicine ,Internal medicine ,Testis ,Genetics ,medicine ,Animals ,ddc:576.5 ,Testosterone ,Androstenedione ,Molecular Biology ,Research Articles ,Mice, Knockout ,chemistry.chemical_classification ,Sertoli Cells ,Testosterone (patch) ,Sertoli cell ,Phenotype ,Mice, Inbred C57BL ,Fertility ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Enzyme ,chemistry ,Androgens ,Female ,030217 neurology & neurosurgery ,Research Article ,Biotechnology - Abstract
Male development, fertility, and lifelong health are all androgen‐dependent. Approximately 95% of circulating testosterone is synthesized by the testis and the final step in this canonical pathway is controlled by the activity of the hydroxysteroid‐dehydrogenase‐17‐beta‐3 (HSD17B3). To determine the role of HSD17B3 in testosterone production and androgenization during male development and function we have characterized a mouse model lacking HSD17B3. The data reveal that developmental masculinization and fertility are normal in mutant males. Ablation of HSD17B3 inhibits hyperstimulation of testosterone production by hCG, although basal testosterone levels are maintained despite the absence of HSD17B3. Reintroduction of HSD17B3 via gene‐delivery to Sertoli cells in adulthood partially rescues the adult phenotype, showing that, as in development, different cell‐types in the testis are able to work together to produce testosterone. Together, these data show that HS17B3 acts as a rate‐limiting‐step for the maximum level of testosterone production by the testis but does not control basal testosterone production. Measurement of other enzymes able to convert androstenedione to testosterone identifies HSD17B12 as a candidate enzyme capable of driving basal testosterone production in the testis. Together, these findings expand our understanding of testosterone production in males.
- Published
- 2020
28. Exogenous Gonadotrophin Stimulation Induces Partial Maturation of Human Sertoli Cells in a Testicular Xenotransplantation Model for Fertility Preservation
- Author
-
Marsida Hutka, Ellen Goossens, W. Hamish B. Wallace, Rod T. Mitchell, Lee B. Smith, Jan-Bernd Stukenborg, Basic (bio-) Medical Sciences, and Biology of the Testis
- Subjects
Sertoli cell ,Leydig cell ,steroidogenesis ,hCG ,human fetal testis ,xenotransplantation ,fertility preservation ,oncofertility ,endocrine system ,sertoli cell ,Somatic cell ,Xenotransplantation ,medicine.medical_treatment ,hcg ,lcsh:Medicine ,Article ,Human chorionic gonadotropin ,Andrology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Medicine(all) ,030219 obstetrics & reproductive medicine ,business.industry ,urogenital system ,lcsh:R ,General Medicine ,leydig cell ,3. Good health ,Transplantation ,Androgen receptor ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,business ,Spermatogenesis - Abstract
The future fertility of prepubertal boys with cancer may be irreversibly compromised by chemotherapy and/or radiotherapy. Successful spermatogenesis has not been achieved following the xenotransplantation of prepubertal human testis tissue, which is likely due to the failure of somatic cell maturation and function. We used a validated xenograft model to identify the factors required for Leydig and Sertoli cell development and function in immature human testis. Importantly, we compared the maturation status of Sertoli cells in xenografts with that of human testis tissues (n = 9, 1 year-adult). Human fetal testis (n = 6; 14−21 gestational weeks) tissue, which models many aspects of prepubertal testicular development, was transplanted subcutaneously into castrated immunocompromised mice for ~12 months. The mice received exogenous human chorionic gonadotropin (hCG; 20IU, 3×/week). In xenografts exposed continuously to hCG, we demonstrate the maintenance of Leydig cell steroidogenesis, the acquisition of features of Sertoli cell maturation (androgen receptor, lumen development), and the formation of the blood−testis barrier (connexin 43), none of which were present prior to the transplantation or in xenografts in which hCG was withdrawn after 7 months. These studies provide evidence that hCG plays a role in Sertoli cell maturation, which is relevant for future investigations, helping them generate functional gametes from immature testis tissue for clinical application.
- Published
- 2020
29. Bi-allelic Recessive Loss-of-Function Variants in FANCM Cause Non-obstructive Azoospermia
- Author
-
Alexandra M. Lopes, Eve Laasik, Lee B. Smith, Donald F. Conrad, Margus Punab, Filipa Carvalho, Marina Grigorova, Kenneth I. Aston, Laura Kasak, Ave Minajeva, Anna Maria Punab, Maris Laan, and Liina Nagirnaja
- Subjects
Adult ,Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Loss of Heterozygosity ,Breast Neoplasms ,Biology ,Compound heterozygosity ,Article ,Frameshift mutation ,Sertoli cell-only syndrome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Fanconi anemia ,hemic and lymphatic diseases ,Testis ,Exome Sequencing ,Genetics ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Gene Silencing ,FANCM ,Frameshift Mutation ,Genetics (clinical) ,Exome sequencing ,Azoospermia ,Ovarian Neoplasms ,030219 obstetrics & reproductive medicine ,Homozygote ,DNA Helicases ,Middle Aged ,medicine.disease ,Spermatozoa ,Pedigree ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Codon, Nonsense ,Female ,Germ cell - Abstract
Infertility affects around 7% of men worldwide. Idiopathic non-obstructive azoospermia (NOA) is defined as the absence of spermatozoa in the ejaculate due to failed spermatogenesis. There is a high probability that NOA is caused by rare genetic defects. In this study, whole-exome sequencing (WES) was applied to two Estonian brothers diagnosed with NOA and Sertoli cell-only syndrome (SCOS). Compound heterozygous loss-of-function (LoF) variants in FANCM (Fanconi anemia complementation group M) were detected as the most likely cause for their condition. A rare maternally inherited frameshift variant p.Gln498Thrfs∗7 (rs761250416) and a previously undescribed splicing variant (c.4387−10A>G) derived from the father introduce a premature STOP codon leading to a truncated protein. FANCM exhibits enhanced testicular expression. In control subjects, immunohistochemical staining localized FANCM to the Sertoli and spermatogenic cells of seminiferous tubules with increasing intensity through germ cell development. This is consistent with its role in maintaining genomic stability in meiosis and mitosis. In the individual with SCOS carrying bi-allelic FANCM LoF variants, none or only faint expression was detected in the Sertoli cells. As further evidence, we detected two additional NOA-affected case subjects with independent FANCM homozygous nonsense variants, one from Estonia (p.Gln1701∗; rs147021911) and another from Portugal (p.Arg1931∗; rs144567652). The study convincingly demonstrates that bi-allelic recessive LoF variants in FANCM cause azoospermia. FANCM pathogenic variants have also been linked with doubled risk of familial breast and ovarian cancer, providing an example mechanism for the association between infertility and cancer risk, supported by published data on Fancm mutant mouse models.
- Published
- 2018
30. Deliverable transgenics & gene therapy possibilities for the testes
- Author
-
Lee B. Smith and Annalucia Darbey
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Genetic enhancement ,Biochemistry ,Male infertility ,03 medical and health sciences ,Endocrinology ,Genome editing ,Deliverable ,Testis ,Animals ,Humans ,Medicine ,Transgenes ,Intensive care medicine ,Molecular Biology ,business.industry ,Gene Transfer Techniques ,Treatment options ,Genetic Therapy ,medicine.disease ,030104 developmental biology ,Increased risk ,Male reproductive disorders ,business - Abstract
Male infertility and hypogonadism are clinically prevalent conditions with a high socioeconomic burden and are both linked to an increased risk in cardiovascular-metabolic diseases and earlier mortality. Therefore, there is an urgent need to better understand the causes and develop new treatments for these conditions that affect millions of men. The accelerating advancement in gene editing and delivery technologies promises improvements in both diagnosis as well as affording the opportunity to develop bespoke treatment options which would both prove beneficial for the millions of individuals afflicted with these reproductive disorders. In this review, we summarise the systems developed and utilised for the delivery of gene therapy and discuss how each of these systems could be applied for the development of a gene therapy system in the testis and how they could be of use for the future diagnosis and repair of common male reproductive disorders.
- Published
- 2018
31. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells
- Author
-
Richard J. Griffeth, Isabelle Stévant, François P. Pralong, Jean-Luc Pitetti, Françoise Kühne, Meng-Chun Hu, Silvana A. Andric, Yasmine Neirijnck, Lee B. Smith, Serge Nef, Karen R. Kilcoyne, Pierre Calvel, Université de Genève (UNIGE), Génétique Animale et Biologie Intégrative (GABI), AgroParisTech-Institut National de la Recherche Agronomique (INRA), Université Paris Saclay (COmUE), University of Edinburgh, University of Novi Sad, National Taiwan University, Department of Internal Medicine, University Hospital, School of Environmental and Life Sciences, and Newcastle University [Newcastle]
- Subjects
0301 basic medicine ,Male ,steroidogenesis ,medicine.medical_treatment ,Cellular differentiation ,[SDV]Life Sciences [q-bio] ,MOUSE ,Biochemistry ,testis development ,Mice ,0302 clinical medicine ,ddc:576.5 ,Testosterone ,Receptor ,Mice, Knockout ,TRANSGENIC MICE ,Leydig cell ,TESTIS ,Stem Cells ,PROLIFERATION ,Leydig Cells ,Cell Differentiation ,ADRENAL-GLAND ,medicine.anatomical_structure ,DIFFERENTIATION ,Biotechnology ,EXPRESSION ,medicine.medical_specialty ,endocrine system ,Biology ,MATURATION ,03 medical and health sciences ,Growth factor receptor ,progenitor Leydig cells ,Internal medicine ,GROWTH-FACTOR-I ,Genetics ,medicine ,Animals ,IGF ,Molecular Biology ,Insulin-like growth factor 1 receptor ,adrenal gland ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Insulin ,Receptors, Somatomedin ,Receptor, Insulin ,Insulin receptor ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,030104 developmental biology ,Endocrinology ,PROGENITOR CELLS ,biology.protein ,Adrenal Cortex ,Corticosterone ,030217 neurology & neurosurgery - Abstract
The insulin family of growth factors (insulin, IGF1, and IGF2) are critical in sex determination, adrenal differentiation, and testicular function. Notably, the IGF system has been reported to mediate the proliferation of steroidogenic cells. However, the precise role and contribution of the membrane receptors mediating those effects, namely, insulin receptor (INSR) and type-I insulin-like growth factor receptor (IGF1R), have not, to our knowledge, been investigated. We show here that specific deletion of both Insr and Igf1r in steroidogenic cells in mice leads to severe alterations of adrenocortical and testicular development. Double-mutant mice display drastic size reduction of both adrenocortex and testes, with impaired corticosterone, testosterone, and sperm production. Detailed developmental analysis of the testes revealed that fetal Leydig cell (LC) function is normal, but there is a failure of adult LC maturation and steroidogenic function associated with accumulation of progenitor LCs (PLCs). Cell-lineage tracing revealed PLC enrichment is secondary to Insr and Igf1r deletion in differentiated adult LCs, suggesting a feedback mechanism between cells at different steps of differentiation. Taken together, these data reveal the cell-autonomous and nonautonomous roles of the IGF system for proper development and maintenance of steroidogenic lineages.-Neirijnck, Y., Calvel, P., Kilcoyne, K. R., Kühne, F., Stévant, I., Griffeth, R. J., Pitetti, J.-L., Andric, S. A., Hu, M.-C., Pralong, F., Smith, L. B., Nef, S. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells.
- Published
- 2018
32. LKB1 is an essential regulator of spermatozoa release during spermiation in the mammalian testis.
- Author
-
Fiona C Denison, Lee B Smith, Phillip J Muckett, Laura O'Hara, David Carling, and Angela Woods
- Subjects
Medicine ,Science - Abstract
LKB1 acts as a master upstream protein kinase regulating a number of kinases involved in diverse cellular functions. Recent studies have suggested a role for LKB1 in male fertility. Male mice with reduced total LKB1 expression, including the complete absence of the major splice variant in testis (LKB1(S)), are completely infertile. We sought to further characterise these mice and determine the mechanism underlying this infertility. This involved expression studies of LKB1 in developing germ cells, morphological analysis of mature spermatozoa and histological studies of both the testis and epididymis using light microscopy and transmission electron microscopy. We conclude that a defect in the release of mature spermatids from the seminiferous epithelium (spermiation) during spermatozoan development is a major cause of the infertility phenotype. We also present evidence that this is due, at least in part, to defects in the breakdown of the junctions, known as ectoplasmic specialisations, between the sertoli cells of the testis epithelium and the heads of the maturing spermatids. Overall this study uncovers a critical role for LKB1 in spermiation, a highly regulated, but poorly understood process vital for male fertility.
- Published
- 2011
- Full Text
- View/download PDF
33. Xenotransplantation as a model for human testicular development
- Author
-
Lee B. Smith, Marsida Hutka, and Rod T. Mitchell
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Sex Differentiation ,Xenotransplantation ,medicine.medical_treatment ,media_common.quotation_subject ,Transplantation, Heterologous ,Embryonic Development ,Physiology ,Fertility ,Disease ,Endocrine Disruptors ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Prepuberty ,Testis ,medicine ,Animals ,Humans ,Fertility preservation ,Molecular Biology ,media_common ,030219 obstetrics & reproductive medicine ,Cancer ,Environmental Exposure ,Cell Biology ,Environmental exposure ,medicine.disease ,Transplantation ,030104 developmental biology ,Immunology ,Developmental Biology - Abstract
The developing male reproductive system may be sensitive to disruption by a wide range of exogenous 'endocrine disruptors'. In-utero exposure to environmental chemicals and pharmaceuticals have been hypothesized to have an impact in the increasing incidence of male reproductive disorders. The vulnerability to adverse effects as a consequence of such exposures is elevated during a specific 'window of susceptibility' in fetal life referred to as the masculinisation programing window (MPW). Exposures that occur during prepuberty, such as chemotherapy treatment for cancer during childhood, may also affect future fertility. Much of our current knowledge about fetal and early postnatal human testicular development derives from studies conducted in animal models predictive for humans. Therefore, over recent years, testicular transplantation has been employed as a 'direct' approach to understand the development of human fetal and prepubertal testis in health and disease. In this review we describe the potential use of human testis xenotransplantation to study testicular development and its application for (i) assessing the effects of environmental exposures in humans, and (ii) establishing fertility preservation options for prepubertal boys with cancer.
- Published
- 2017
34. A missense mutation in Katnal1 underlies behavioural, neurological and ciliary anomalies
- Author
-
Petrina Lau, Michelle Simon, Anna Hoerder-Suabedissen, Federico Tinarelli, Andrew Rutman, Patrick M. Nolan, Neil R. Horner, Matthew Sweeting, Johanna E. Chesham, Sara Johnson, Henrik Westerberg, Gareth Banks, Ashleigh G. Wilcox, Zoltán Molnár, Alun R. Barnard, Valter Tucci, Robert A. Hirst, Glenda Lassi, Lee B. Smith, Michael H. Hastings, and Thomas N. Lawson
- Subjects
0301 basic medicine ,Microcephaly ,Mutation, Missense ,Katanin ,Microtubules ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,Ependyma ,medicine ,Missense mutation ,Animals ,Humans ,Cilia ,Allele ,Molecular Biology ,Loss function ,Microtubule severing ,Adenosine Triphosphatases ,Neurons ,biology ,medicine.disease ,Phenotype ,Circadian Rhythm ,Mice, Inbred C57BL ,Psychiatry and Mental health ,030104 developmental biology ,Mutation ,Motile cilium ,biology.protein ,Original Article ,Psychology ,Sleep ,Neuroscience - Abstract
Microtubule severing enzymes implement a diverse range of tissue-specific molecular functions throughout development and into adulthood. Although microtubule severing is fundamental to many dynamic neural processes, little is known regarding the role of the family member Katanin p60 subunit A-like 1, KATNAL1, in central nervous system (CNS) function. Recent studies reporting that microdeletions incorporating the KATNAL1 locus in humans result in intellectual disability and microcephaly suggest that KATNAL1 may play a prominent role in the CNS; however, such associations lack the functional data required to highlight potential mechanisms which link the gene to disease symptoms. Here we identify and characterise a mouse line carrying a loss of function allele in Katnal1. We show that mutants express behavioural deficits including in circadian rhythms, sleep, anxiety and learning/memory. Furthermore, in the brains of Katnal1 mutant mice we reveal numerous morphological abnormalities and defects in neuronal migration and morphology. Furthermore we demonstrate defects in the motile cilia of the ventricular ependymal cells of mutants, suggesting a role for Katnal1 in the development of ciliary function. We believe the data we present here are the first to associate KATNAL1 with such phenotypes, demonstrating that the protein plays keys roles in a number of processes integral to the development of neuronal function and behaviour.Molecular Psychiatry advance online publication, 4 April 2017; doi:10.1038/mp.2017.54.
- Published
- 2017
- Full Text
- View/download PDF
35. Human Adipose-derived Pericytes Display Steroidogenic Lineage Potential in Vitro and Influence Leydig Cell Regeneration in Vivo in Rats
- Author
-
Laura Milne, Ian Handel, Lee B. Smith, Michael Curley, Z. Gonzalez, Patrick W. F. Hadoke, and Bruno Péault
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Adipose tissue ,lcsh:Medicine ,Reproductive biology ,Cell Count ,Biology ,Rats, Inbred WKY ,Article ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Internal medicine ,Testis ,medicine ,Regeneration ,Animals ,Humans ,Cell Lineage ,RNA, Messenger ,lcsh:Science ,Testosterone ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,030219 obstetrics & reproductive medicine ,Multidisciplinary ,Leydig cell ,Regeneration (biology) ,lcsh:R ,Leydig Cells ,Organ Size ,Androgen ,Hormones ,Transplantation ,medicine.anatomical_structure ,Endocrinology ,Adipose Tissue ,Gene Expression Regulation ,lcsh:Q ,Steroids ,Stem cell ,Pericytes - Abstract
Exogenous androgen replacement is used to treat symptoms associated with low testosterone in males. However, adverse cardiovascular risk and negative fertility impacts impel development of alternative approaches to restore/maintain Leydig cell (LC) androgen production. Stem Leydig cell (SLC) transplantation shows promise in this regard however, practicality of SLC isolation/transplantation impede clinical translation. Multipotent human adipose-derived perivascular stem cells (hAd-PSCs) represent an attractive extragonadal stem cell source for regenerative therapies in the testis but their therapeutic potential in this context is unexplored. We asked whether hAd-PSCs could be converted into Leydig-like cells and determined their capacity to promote regeneration in LC-ablated rat testes. Exposure of hAd-PSCs to differentiation-inducing factors in vitro upregulated steroidogenic genes but did not fully induce LC differentiation. In vivo, no difference in LC-regeneration was noted between Sham and hAd-PSC-transplanted rats. Interestingly, Cyp17a1 expression increased in hAd-PSC-transplanted testes compared to intact vehicle controls and the luteinising hormone/testosterone ratio returned to Vehicle control levels which was not the case in EDS + Sham animals. Notably, hAd-PSCs were undetectable one-month after transplantation suggesting this effect is likely mediated via paracrine mechanisms during the initial stages of regeneration; either directly by interacting with regenerating LCs, or through indirect interactions with trophic macrophages.
- Published
- 2019
36. Androgen receptor signalling in the male adrenal facilitates X-zone regression, cell turnover and protects against adrenal degeneration during ageing
- Author
-
Laura Milne, Rod T. Mitchell, Lee B. Smith, Anne Jørgensen, Sarah Smith, Hanne Frederiksen, Anne-Louise Gannon, J. Ian Mason, and Laura O’Hara
- Subjects
Male ,0301 basic medicine ,Aging ,medicine.medical_specialty ,medicine.drug_class ,Cell ,lcsh:Medicine ,Apoptosis ,Biology ,Protective Agents ,urologic and male genital diseases ,Article ,Adrenal tumours ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Cortex (anatomy) ,medicine ,Animals ,Humans ,Castration ,lcsh:Science ,Receptor ,Mice, Knockout ,Adrenal gland diseases ,Multidisciplinary ,Adrenal cortex ,lcsh:R ,Hyperplasia ,Androgen ,medicine.disease ,Mice, Inbred C57BL ,Androgen receptor ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Receptors, Androgen ,Adrenal Cortex ,Androgens ,lcsh:Q ,Signal transduction ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Androgens are known to be an essential regulator of male health. Androgen receptor (AR) is widely expressed throughout the adrenal cortex, yet the wider role for androgen signalling in the adrenal remains underexplored. To investigate AR-dependent and AR-independent androgen signalling in the adrenal, we used a novel mouse model with a specific ablation of androgen receptor in the adrenal cortex with or without reduction of circulating androgen levels by castration. Our results describe AR expression in the human and mouse adrenal and highlight that the mouse is a viable model to investigate androgen signalling in the adrenal cortex. We show androgen signalling via AR is required for X-zone regression during puberty. Furthermore, cortex measurements define differences in X-zone morphology depending on whether circulating androgens or AR have been removed. We show androgens promote both cortical cell differentiation and apoptosis but are dispensable for the formation of the definitive cortex. Additionally, investigation of aged mice with AR ablation reveals severe cortex disruption, spindle cell hyperplasia and X-zone expansion. The data described herein demonstrates AR-signalling is required to facilitate X-zone regression, cell clearance and to protect against adrenal degeneration during ageing.
- Published
- 2019
37. Dental Care Needs of Male versus Female Children Visiting a School-based Mobile Dental Facility in West Virginia
- Author
-
R Constance, Wiener, Tiffany, Summerlin, Lee B, Smith, Daniel T, Carrier, and Michael A, Wiener
- Subjects
Male ,Dental Facilities ,Humans ,Female ,Oral Health ,Dental Caries ,West Virginia ,Child ,Dental Care - Published
- 2019
38. Ablation of glucocorticoid receptor in the hindbrain of the mouse provides a novel model to investigate stress disorders
- Author
-
Anne Jørgensen, Adriana Traveres, Hanne Frederiksen, Lee B. Smith, Anne-Louise Gannon, Carlos J. Alcaide-Corral, Sarah Smith, Rod T. Mitchell, J. Ian Mason, Diane Rebourcet, Laura O’Hara, and Laura Milne
- Subjects
0301 basic medicine ,Male ,lcsh:Medicine ,Hindbrain ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Glucocorticoid receptor ,Receptors, Glucocorticoid ,Corticosterone ,medicine ,Animals ,Kyphosis ,Receptor ,lcsh:Science ,Mice, Knockout ,Recombination, Genetic ,Multidisciplinary ,Behavior, Animal ,Adrenal cortex ,business.industry ,lcsh:R ,Body Weight ,Mice, Inbred C57BL ,Rhombencephalon ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Positron-Emission Tomography ,Forebrain ,Adrenal Cortex ,Anxiety ,lcsh:Q ,Female ,medicine.symptom ,business ,Tomography, X-Ray Computed ,Neuroscience ,030217 neurology & neurosurgery ,Glucocorticoid ,Stress, Psychological ,medicine.drug - Abstract
The hypothalamic-pituitary-adrenal (HPA) axis regulates responses to internal and external stressors. Many patients diagnosed with conditions such as depression or anxiety also have hyperactivity of the HPA axis. Hyper-stimulation of the HPA axis results in sustained elevated levels of glucocorticoids which impair neuronal function and can ultimately result in a psychiatric disorder. Studies investigating Glucocorticoid Receptor (GR/NR3C1) in the brain have primarily focused on the forebrain, however in recent years, the hindbrain has become a region of interest for research into the development of anxiety and depression, though the role of GR signalling in the hindbrain remains poorly characterised. To determine the role of glucocorticoid signalling in the hindbrain we have developed a novel mouse model that specifically ablates hindbrain GR to ascertain its role in behaviour, HPA-axis regulation and adrenal structure. Our study highlights that ablation of GR in the hindbrain results in excessive barbering, obsessive compulsive digging and lack of cage exploration. These mice also develop kyphosis, elevated circulating corticosterone and severe adrenal cortex disruption. Together, this data demonstrates a role for hindbrain GR signalling in regulating stress-related behaviour and identifies a novel mouse model to allow further investigation into the pathways impacting stress and anxiety.
- Published
- 2019
39. Fertility Preservation in Childhood Cancer: Endocrine Activity in Prepubertal Human Testis Xenografts Exposed to a Pubertal Hormone Environment
- Author
-
Marsida Hutka, Ellen Goossens, Natalie Z.M. Homer, Lee B. Smith, Rod T. Mitchell, Dorien Van Saen, Prashant Kadam, Jaime Onofre, Jan-Bernd Stukenborg, W. Hamish B. Wallace, Basic (bio-) Medical Sciences, and Biology of the Testis
- Subjects
0301 basic medicine ,steroidogenesis ,endocrine system ,Cancer Research ,fertility preservation ,hCG ,gonadotoxicity ,Biology ,prepubertal human testis ,lcsh:RC254-282 ,Article ,Andrology ,03 medical and health sciences ,0302 clinical medicine ,Seminal vesicle ,xenotransplantation ,FSH ,medicine ,childhood cancer ,Endocrine system ,Fertility preservation ,030219 obstetrics & reproductive medicine ,Cholesterol side-chain cleavage enzyme ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Sertoli cell ,Transplantation ,side effects ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,testosterone ,Germ cell ,Hormone - Abstract
Survivors of childhood cancer are at risk for long-term treatment-induced health sequelae, including gonadotoxicity and iatrogenic infertility. At present, for prepubertal boys there are no viable clinical options to preserve future reproductive potential. We investigated the effect of a pubertal induction regimen with gonadotrophins on prepubertal human testis xenograft development. Human testis tissue was obtained from patients with cancer and non-malignant haematological disorders (n = 6, aged 1&ndash, 14 years) who underwent testis tissue cryopreservation for fertility preservation. Fresh and frozen-thawed testis fragments were transplanted subcutaneously or intratesticularly into immunocompromised mice. Graft-bearing mice received injections of vehicle or exogenous gonadotrophins, human chorionic gonadotrophin (hCG, 20 IU), and follicle-stimulating hormone (FSH, 12.5 IU) three times a week for 12 weeks. The gross morphology of vehicle and gonadotrophin-exposed grafts was similar for both transplantation sites. Exposure of prepubertal human testis tissue xenografts to exogenous gonadotrophins resulted in limited endocrine function of grafts, as demonstrated by the occasional expression of the steroidogenic cholesterol side-chain cleavage enzyme (CYP11A1). Plasma testosterone concentrations (0.13 vs. 0.25 ng/mL, p = 0.594) and seminal vesicle weights (10.02 vs. 13.93 mg, p = 0.431) in gonadotrophin-exposed recipient mice were comparable to vehicle-exposed controls. Regardless of the transplantation site and treatment, initiation and maintenance of androgen receptor (AR) expression were observed in Sertoli cells, indicating commitment towards a more differentiated status. However, neither exogenous gonadotrophins (in castrated host mice) nor endogenous testosterone (in intact host mice) were sufficient to repress the expression of markers associated with immature Sertoli cells, such as anti-Mü, llerian hormone (AMH) and Ki67, or to induce the redistribution of junctional proteins (connexin, CX43, claudin 11, CLDN11) to areas adjacent to the basement membrane. Spermatogonia did not progress developmentally but remained the most advanced germ cell type in testis xenografts. Overall, these findings demonstrate that exogenous gonadotrophins promote partial activation and maturation of the somatic environment in prepubertal testis xenografts. However, alternative hormone regimens or additional factors for pubertal induction are required to complete the functional maturation of the spermatogonial stem cell (SSC) niche.
- Published
- 2020
40. A young testicular microenvironment protects Leydig cells against age-related dysfunction in a mouse model of premature aging
- Author
-
Michael, Curley, Laura, Milne, Sarah, Smith, Anne, Jørgensen, Hanne, Frederiksen, Patrick, Hadoke, Paul, Potter, and Lee B, Smith
- Subjects
Male ,Mice, Knockout ,endocrine system ,steroidogenesis ,Research ,Autophagy-Related Proteins ,Leydig Cells ,Aging, Premature ,Nerve Tissue Proteins ,Luteinizing Hormone ,testis ,Mice ,Models, Animal ,testosterone ,Animals ,gonadotropin ,Carrier Proteins ,Gene Deletion - Abstract
Testicular Leydig cells (LCs) are the primary source of circulating androgen in men. As men age, circulating androgen levels decline. However, whether reduced LC steroidogenesis results from specific effects of aging within LCs or reflects degenerative alterations to the wider supporting microenvironment is unclear; inability to separate intrinsic LC aging from that of the testicular microenvironment in vivo has made this question difficult to address. To resolve this, we generated novel mouse models of premature aging, driven by CDGSH iron sulfur domain 2 ( Cisd2) deletion, to separate the effects of cell intrinsic aging from extrinsic effects of aging on LC function. At 6 mo of age, constitutive Cisd2-deficient mice display signs of premature aging, including testicular atrophy, reduced LC and Sertoli cell (SC) number, decreased circulating testosterone, increased luteinizing hormone/testosterone ratio, and decreased expression of steroidogenic mRNAs, appropriately modeling primary testicular dysfunction observed in aging men. However, mice with Cisd2 deletion (and thus premature aging) restricted to either LCs or SCs were protected against testicular degeneration, demonstrating that age-related LCs dysfunction cannot be explained by intrinsic aging within either the LC or SC lineages alone. We conclude that age-related LC dysfunction is largely driven by aging of the supporting testicular microenvironment.-Curley, M., Milne, L., Smith, S., Jørgensen, A., Frederiksen, H., Hadoke, P., Potter, P., Smith, L. B. A Young testicular microenvironment protects Leydig cells against age-related dysfunction in a mouse model of premature aging.
- Published
- 2018
41. Leukemia Inhibitory Factor-Receptor is Dispensable for Prenatal Testis Development but is Required in Sertoli cells for Normal Spermatogenesis in Mice
- Author
-
Michael Curley, Laura Milne, Sarah Smith, Nina Atanassova, Diane Rebourcet, Annalucia Darbey, Patrick W. F. Hadoke, Sara Wells, and Lee B. Smith
- Subjects
Male ,Mice, Knockout ,TRANSGENIC MICE ,GERM-CELLS ,endocrine system ,Sertoli Cells ,LIF RECEPTOR ,Leukemia Inhibitory Factor Receptor alpha Subunit ,urogenital system ,lcsh:R ,ANDROGEN RECEPTOR ,GP130 ,lcsh:Medicine ,Article ,Mice ,RAT SERTOLI ,IL-6 SIGNAL TRANSDUCER ,Testis ,CRE RECOMBINASE ACTIVITY ,MALE REPRODUCTIVE-SYSTEM ,Animals ,lcsh:Q ,LEYDIG-CELLS ,lcsh:Science ,Spermatogenesis - Abstract
Leukemia inhibitory factor (LIF), a pleiotropic cytokine belonging to the interleukin-6 family, is most often noted for its role in maintaining the balance between stem cell proliferation and differentiation. In rodents, LIF is expressed in both the fetal and adult testis; with the peritubular myoid (PTM) cells thought to be the main site of production. Given their anatomical location, LIF produced by PTM cells may act both on intratubular and interstitial cells to influence spermatogenesis and steroidogenesis respectively. Indeed, the leukemia inhibitory factor receptor (LIFR) is expressed in germ cells, Sertoli cells, Leydig cells, PTM cells and testicular macrophages, suggesting that LIF signalling via LIFR may be a key paracrine regulator of testicular function. However, a precise role(s) for testicular LIFR-signalling in vivo has not been established. To this end, we generated and characterised the testicular phenotype of mice lacking LIFR either in germ cells, Sertoli cells or both, to identify a role for LIFR-signalling in testicular development/function. Our analyses reveal that LIFR is dispensable in germ cells for normal spermatogenesis. However, Sertoli cell LIFR ablation results in a degenerative phenotype, characterised by abnormal germ cell loss, sperm stasis, seminiferous tubule distention and subsequent atrophy of the seminiferous tubules.
- Published
- 2018
42. DMRT1 repression using a novel approach to genetic manipulation induces testicular dysgenesis in human fetal gonads
- Author
-
Joni, Macdonald, Karen R, Kilcoyne, Richard M, Sharpe, Áine, Kavanagh, Richard A, Anderson, Pamela, Brown, Lee B, Smith, Anne, Jørgensen, and Rod T, Mitchell
- Subjects
Male ,endocrine system ,Reproductive Biology ,Sertoli Cells ,gonadal development ,Down-Regulation ,Mice, Nude ,Gonadal Dysgenesis ,DMRT1 ,testicular dysgenesis ,Mice ,MicroRNAs ,Gene Knockdown Techniques ,Testis ,Animals ,Humans ,sex differentiation ,Original Article ,human fetal testis ,Transcription Factors ,miRNA - Abstract
STUDY QUESTION Does loss of DMRT1 in human fetal testis alter testicular development and result in testicular dysgenesis? SUMMARY ANSWER DMRT1 repression in human fetal testis alters the expression of key testicular and ovarian determining genes, and leads to focal testicular dysgenesis. WHAT IS KNOWN ALREADY Testicular dysgenesis syndrome (TDS) is associated with common testicular disorders in young men, but its etiology is unknown. DMRT1 has been shown to play a role in the regulation of sex differentiation in the vertebrate gonad. Downregulation of DMRT1 in male mice results in trans-differentiation of Sertoli cells into granulosa (FOXL2+) cells resulting in an ovarian gonadal phenotype. STUDY DESIGN, SIZE, DURATION To determine the effect of DMRT1 repression on human fetal testes, we developed a novel system for genetic manipulation, which utilizes a Lentivral delivered miRNA during short-term in vitro culture (2 weeks). A long-term (4–6 weeks) ex vivo xenograft model was used to determine the subsequent effects of DMRT1 repression on testicular development and maintenance. We included first and second-trimester testis tissue (8–20 weeks gestation; n = 12) in the study. PARTICIPANTS/MATERIALS, SETTING, METHODS Human fetal testes were cultured in vitro and exposed to either of two DMRT1 miRNAs (miR536, miR641), or to scrambled control miRNA, for 24 h. This was followed by a further 14 days of culture (n = 3–4), or xenografting (n = 5) into immunocompromised mice for 4–6 weeks. Tissues were analyzed by histology, immunohistochemistry, immunofluorescence and quantitative RT-PCR. Endpoints included histological evaluation of seminiferous cord integrity, mRNA expression of testicular, ovarian and germ cell genes, and assessment of cell number and protein expression for proliferation, apoptosis and pluripotency factors. Statistical analysis was performed using a linear mixed effect model. MAIN RESULTS AND THE ROLE OF CHANCE DMRT1 repression (miR536/miR641) resulted in a loss of DMRT1 protein expression in a sub-population of Sertoli cells of first trimester (8–11 weeks gestation) human fetal testis; however, this did not affect the completion of seminiferous cord formation or morphological appearance. In second-trimester testis (12–20 weeks gestation), DMRT1 repression (miR536/miR641) resulted in disruption of seminiferous cords with absence of DMRT1 protein expression in Sertoli (SOX9+) cells. No differences in proliferation (Ki67+) were observed and apoptotic cells (CC3+) were rare. Expression of the Sertoli cell associated gene, SOX8, was significantly reduced (miR536, 34% reduction, P = 0.031; miR641 36% reduction, P = 0.026), whilst SOX9 expression was unaffected. Changes in expression of AMH (miR536, 100% increase, P = 0.033), CYP26B1 (miR641, 38% reduction, P = 0.05) and PTGDS (miR642, 30% reduction, P = 0.0076) were also observed. Amongst granulosa cell associated genes, there was a significant downregulation in R-spondin 1 expression (miR536, 76% reduction, P < 0.0001; miR641, 49% reduction, P = 0.046); however, there were no changes in expression of the granulosa cell marker, FOXL2. Analysis of germ cell associated genes demonstrated a significant increase in the expression of the pluripotency gene OCT4 (miR536, 233%, P < 0.001). We used the xenograft system to investigate the longer-term effects of seminiferous cord disruption via DMRT1 repression. As was evident in vitro for second-trimester samples, DMRT1 repression resulted in focal testicular dysgenesis similar to that described in adults with TDS. These dysgenetic areas were devoid of germ cells, whilst expression of FOXL2 within the dysgenetic areas, indicated trans-differentiation from a male (Sertoli cell) to female (granulosa cell) phenotype. LIMITATIONS, REASONS FOR CAUTION Human fetal testis tissue is a limited resource; however, we were able to demonstrate significant effects of DMRT1 repression on the expression of germ and somatic cell genes, in addition to the induction of focal testicular dysgenesis, using these limited samples. In vitro culture may not reflect all aspects of human fetal testis development and function; however, the concurrent use of the xenograft model which represents a more physiological system supports the validity of the in vitro findings. WIDER IMPLICATIONS OF THE FINDINGS Our findings have important implications for understanding the role of DMRT1 in human testis development and in the origin of testicular dysgenesis. In addition, we provide validation of a novel system that can be used to determine the effects of repression of genes that have been implicated in gonadal development and associated human reproductive disorders. STUDY FUNDING/COMPETING INTEREST(S) This project was funded by a Wellcome Trust Intermediate Clinical Fellowship (Grant No. 098522) awarded to RTM. LBS was supported by MRC Programme Grant MR/N002970/1. RAA was supported by MRC Programme Grant G1100357/1. RMS was supported by MRC Programme Grant G33253. This work was undertaken in the MRC Centre for Reproductive Health which is funded by the MRC Centre grant MR/N022556/1. The funding bodies had no input into the conduct of the research or the production of this manuscript. The authors have declared no conflicts of interest.
- Published
- 2018
43. A graphical and computational modeling platform for biological pathways
- Author
-
Alessandra Livigni, Tom C. Freeman, Laura O'Hara, Marta E Polak, Derek W. Wright, Tim Angus, and Lee B. Smith
- Subjects
0301 basic medicine ,Scheme (programming language) ,Theoretical computer science ,Computer science ,0206 medical engineering ,02 engineering and technology ,Notation ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Software ,Component (UML) ,Journal Article ,Computer Graphics ,Computer Simulation ,Protein Interaction Maps ,computer.programming_language ,Computational model ,business.industry ,Systems Biology ,Computational Biology ,Petri net ,Visualization ,030104 developmental biology ,business ,computer ,020602 bioinformatics ,Algorithms ,Metabolic Networks and Pathways ,Network analysis - Abstract
This biologist-friendly modeling scheme facilitates the capture and visualization of knowledge about biological pathways and how components interact. These pathway models can be directly used to run simulations of their activity and test hypotheses. A major endeavor of systems biology is the construction of graphical and computational models of biological pathways as a means to better understand their structure and function. Here, we present a protocol for a biologist-friendly graphical modeling scheme that facilitates the construction of detailed network diagrams, summarizing the components of a biological pathway (such as proteins and biochemicals) and illustrating how they interact. These diagrams can then be used to simulate activity flow through a pathway, thereby modeling its dynamic behavior. The protocol is divided into four sections: (i) assembly of network diagrams using the modified Edinburgh Pathway Notation (mEPN) scheme and yEd network editing software with pathway information obtained from published literature and databases of molecular interaction data; (ii) parameterization of the pathway model within yEd through the placement of 'tokens' on the basis of the known or imputed amount or activity of a component; (iii) model testing through visualization and quantitative analysis of the movement of tokens through the pathway, using the network analysis tool Graphia Professional and (iv) optimization of model parameterization and experimentation. This is the first modeling approach that combines a sophisticated notation scheme for depicting biological events at the molecular level with a Petri net–based flow simulation algorithm and a powerful visualization engine with which to observe the dynamics of the system being modeled. Unlike many mathematical approaches to modeling pathways, it does not require the construction of a series of equations or rate constants for model parameterization. Depending on a model's complexity and the availability of information, its construction can take days to months, and, with refinement, possibly years. However, once assembled and parameterized, a simulation run, even on a large model, typically takes only seconds. Models constructed using this approach provide a means of knowledge management, information exchange and, through the computation simulation of their dynamic activity, generation and testing of hypotheses, as well as prediction of a system's behavior when perturbed.
- Published
- 2018
44. Testicular Cell Selective Ablation Using Diphtheria Toxin Receptor Transgenic Mice
- Author
-
Diane, Rebourcet, Annalucia, Darbey, Michael, Curley, Peter, O'Shaughnessy, and Lee B, Smith
- Subjects
Male ,Mice ,Germ Cells ,Sertoli Cells ,Animals ,Diphtheria Toxin ,Mice, Transgenic ,Spermatogenesis ,Cells, Cultured ,Poisons ,Heparin-binding EGF-like Growth Factor - Abstract
Testis development and function is regulated by intricate cell-cell cross talk. Characterization of the mechanisms underpinning this has been derived through a wide variety of approaches including pharmacological manipulation, transgenics, and cell-specific ablation of populations. The removal of all or a proportion of a specific cell type has been achieved through a variety of approaches. In this paper, we detail a combined transgenic and pharmacological approach to ablate the Sertoli or germ cell populations using diphtheria toxin in mice. We describe the key steps in generation, validation, and use of the models and also describe the caveats and cautions necessary. We also provide a detailed description of the methodology applied to characterize testis development and function in models of postnatal Sertoli or germ cell ablation.
- Published
- 2018
45. Androgen Receptor
- Author
-
Iain J. McEwan and Lee B. Smith
- Published
- 2018
46. Testicular Cell Selective Ablation Using Diphtheria Toxin Receptor Transgenic Mice
- Author
-
Lee B. Smith, Annalucia Darbey, Diane Rebourcet, Michael Curley, and Peter J. O'Shaughnessy
- Subjects
0301 basic medicine ,Genetically modified mouse ,Diphtheria toxin ,030219 obstetrics & reproductive medicine ,Selective ablation ,Transgene ,Cell ,Biology ,Sertoli cell ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Germ cell ,Function (biology) - Abstract
Testis development and function is regulated by intricate cell-cell cross talk. Characterization of the mechanisms underpinning this has been derived through a wide variety of approaches including pharmacological manipulation, transgenics, and cell-specific ablation of populations. The removal of all or a proportion of a specific cell type has been achieved through a variety of approaches. In this paper, we detail a combined transgenic and pharmacological approach to ablate the Sertoli or germ cell populations using diphtheria toxin in mice. We describe the key steps in generation, validation, and use of the models and also describe the caveats and cautions necessary. We also provide a detailed description of the methodology applied to characterize testis development and function in models of postnatal Sertoli or germ cell ablation.
- Published
- 2018
47. Correction: Pleiotropic Impacts of Macrophage and Microglial Deficiency on Development in Rats with Targeted Mutation of the Csf1r Locus
- Author
-
Lucas Lefevre, Kim M. Summers, Tom Burdon, Alison J. Thomson, Kyle R. Upton, Gemma M. Davis, Rocio Rojo, Kristin A. Sauter, Neil A. Mabbott, Joana Alves, Lee B. Smith, Yi Ting Tsai, Kathleen Grabert, Robert Wallace, Anna Raper, Tim Regan, David A. Hume, Sara Clohisey, Pedro Piccardo, Stephen Meek, Zofia M. Lisowski, Michael Cheeseman, Clare Pridans, Stephen J. Bush, Deborah Brown, and Linda Sutherland
- Subjects
Genetics ,Pituitary gland ,Immunology ,Locus (genetics) ,Striatum ,Biology ,Olfactory bulb ,medicine.anatomical_structure ,Targeted Mutation ,Genotype ,Gene expression ,medicine ,Immunology and Allergy ,Gene - Abstract
In the analysis of gene expression in the brain, the sample labels for the pituitary gland and olfactory bulb were reversed. As a consequence, the clusters in Fig. 10B were mislabeled. A corrected version of Fig. 10 is shown below, along with the corrected figure legend. These corrections have been made to the online version of the article, which now differs from the print version as originally published. A corrected version of Supplemental Table I has also been published online. The current online supplemental material therefore differs from what was originally published online. The relevant text in the Results section under the heading “Network analysis of gene expression in the brain of Csf1r-deficient rats” now reads as follows: Fig. 10B shows the network graph for the combined analysis of the four brain regions; gene lists are provided in Supplemental Table I. The largest cluster was cluster 1 (4283 genes), containing pituitary gland-associated genes. Other region-specific clusters were cluster 3 (olfactory bulb; 878 genes), cluster 5 (hippocampus; 513 genes), and cluster 7 (striatum; 384 genes). The second largest cluster (cluster 2; 1253 genes) contained genes that were more highly expressed in both hippocampus and striatum. Two other main clusters also shared highly expressed genes between tissues: cluster 4 (olfactory bulb, striatum, and hippocampus; 757 genes) and cluster 6 (pituitary gland and olfactory bulb; 471 genes). None of these clusters showed any evidence of genotype association. (Figure Presented).
- Published
- 2019
48. Identification of Sertoli cell-specific transcripts in the mouse testis and the role of FSH and androgen in the control of Sertoli cell activity
- Author
-
Lee B. Smith, Ugo Soffientini, M. H. Abel, G Hamilton, Paul Fowler, Diane Rebourcet, Peter J. O'Shaughnessy, and Se Bee Lee
- Subjects
Male ,0301 basic medicine ,endocrine system ,Sertoli ,lcsh:QH426-470 ,Microarray ,medicine.drug_class ,lcsh:Biotechnology ,Biology ,Androgen ,Transcriptome ,Andrology ,Mice ,03 medical and health sciences ,Follicle-stimulating hormone ,lcsh:TP248.13-248.65 ,Testis ,Genetics ,medicine ,Animals ,Diphtheria Toxin ,Busulfan ,Mice, Knockout ,Sertoli Cells ,urogenital system ,RNAseq ,Sertoli cell ,Spermatozoa ,3. Good health ,Androgen receptor ,lcsh:Genetics ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Androgen ,Hormone receptor ,Androgens ,Receptors, FSH ,Follicle Stimulating Hormone ,Germ cell ,Research Article ,Biotechnology - Abstract
Background The Sertoli cells act to induce testis differentiation and subsequent development in fetal and post-natal life which makes them key to an understanding of testis biology. As a major step towards characterisation of factors involved in Sertoli cell function we have identified Sertoli cell-specific transcripts in the mouse testis and have used the data to identify Sertoli cell-specific transcripts altered in mice lacking follicle-stimulating hormone receptors (FSHRKO) and/or androgen receptors (AR) in the Sertoli cells (SCARKO). Results Adult iDTR mice were injected with busulfan to ablate the germ cells and 50 days later they were treated with diphtheria toxin (DTX) to ablate the Sertoli cells. RNAseq carried out on testes from control, busulfan-treated and busulfan + DTX-treated mice identified 701 Sertoli-specific transcripts and 4302 germ cell-specific transcripts. This data was mapped against results from microarrays using testicular mRNA from 20 day-old FSHRKO, SCARKO and FSHRKO.SCARKO mice. Results show that of the 534 Sertoli cell-specific transcripts present on the gene chips, 85% were altered in the FSHRKO mice and 94% in the SCARKO mice (mostly reduced in both cases). In the FSHRKO.SCARKO mice additive or synergistic effects were seen for most transcripts. Age-dependent studies on a selected number of Sertoli cell-specific transcripts, showed that the marked effects in the FSHRKO at 20 days had largely disappeared by adulthood although synergistic effects of FSHR and AR knockout were seen. Conclusions These studies have identified the Sertoli cell-specific transcriptome in the mouse testis and have shown that most genes in the transcriptome are FSH- and androgen-dependent at puberty although the importance of FSH diminishes towards adulthood. Electronic supplementary material The online version of this article (10.1186/s12864-017-4357-3) contains supplementary material, which is available to authorized users.
- Published
- 2017
49. Cell-specific ablation in the testis: what have we learned?
- Author
-
Diane Rebourcet, Peter J. O'Shaughnessy, and Lee B. Smith
- Subjects
Ablation Techniques ,Male ,germ cells ,Cell type ,Cell signaling ,gonadal development ,Urology ,Endocrinology, Diabetes and Metabolism ,Cell ,Population ,Review Article ,Cell Communication ,germ cell transplantation ,testis ,Biology ,Paracrine signalling ,Endocrinology ,medicine ,Animals ,Humans ,paracrine factors ,Autocrine signalling ,education ,Review Articles ,education.field_of_study ,Sertoli Cells ,Macrophages ,Leydig Cells ,Sertoli cell ,Spermatozoa ,animal models ,spermatogenesis ,Cell biology ,medicine.anatomical_structure ,Reproductive Medicine ,Models, Animal ,Immunology ,Signal transduction ,Signal Transduction - Abstract
Summary Testicular development and function is the culmination of a complex process of autocrine, paracrine and endocrine interactions between multiple cell types. Dissecting this has classically involved the use of systemic treatments to perturb endocrine function, or more recently, transgenic models to knockout individual genes. However, targeting genes one at a time does not capture the more wide‐ranging role of each cell type in its entirety. An often overlooked, but extremely powerful approach to elucidate cellular function is the use of cell ablation strategies, specifically removing one cellular population and examining the resultant impacts on development and function. Cell ablation studies reveal a more holistic overview of cell–cell interactions. This not only identifies important roles for the ablated cell type, which warrant further downstream study, but also, and importantly, reveals functions within the tissue that occur completely independently of the ablated cell type. To date, cell ablation studies in the testis have specifically removed germ cells, Leydig cells, macrophages and recently Sertoli cells. These studies have provided great leaps in understanding not possible via other approaches; as such, cell ablation represents an essential component in the researchers’ tool‐kit, and should be viewed as a complement to the more mainstream approaches to advancing our understanding of testis biology. In this review, we summarise the cell ablation models used in the testis, and discuss what each of these have taught us about testis development and function.
- Published
- 2015
50. Influence of Testosterone on Inflammatory Response in Testicular Cells and Expression of Transcription Factor Foxp3 in T Cells
- Author
-
Laura O’Hara, Ferial Aslani, Florian Eisel, Nelli Baal, Magdalena Walecki, Lee B. Smith, Amir Rafiq, Andreas Meinhardt, Sudhanshu Bhushan, Monika Fijak, Gerhard Schuler, Holger Hackstein, Eva Wahle, Lara Jil Damm, Lutz Konrad, and Jan Per Wenzel
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,medicine.drug_class ,Immunology ,Biology ,urologic and male genital diseases ,T-Lymphocytes, Regulatory ,Flow cytometry ,Immune system ,Transforming Growth Factor beta ,Internal medicine ,medicine ,Animals ,Immunology and Allergy ,Testosterone ,RNA, Messenger ,IL-2 receptor ,Rats, Wistar ,Cells, Cultured ,Chemokine CCL2 ,Inflammation ,Sertoli Cells ,medicine.diagnostic_test ,Tumor Necrosis Factor-alpha ,urogenital system ,Macrophages ,Leydig Cells ,Obstetrics and Gynecology ,FOXP3 ,Androgen Antagonists ,Cell Differentiation ,Forkhead Transcription Factors ,Androgen ,Sertoli cell ,Flutamide ,Interleukin-10 ,Rats ,Endocrinology ,medicine.anatomical_structure ,Reproductive Medicine ,Culture Media, Conditioned ,Tumor necrosis factor alpha - Abstract
Problem Previous studies demonstrated a strong association between low androgen levels and reduced capacity to mount an inflammatory response. However, the mechanisms underlying these observations are largely not understood. Methods of study Generation of CD4+CD25+Foxp3+ regulatory T cells in Leydig cell-conditioned media was determined by flow cytometry and ELISA. Influence of testosterone on cytokine response was measured in LPS-stimulated testicular macrophages, Sertoli and peritubular cells. Results Leydig cell-conditioned media dose-dependently stimulated expression of transcription factor Foxp3 and secretion of IL-10 in splenic CD4+ T cells, an effect abolished by addition of the anti-androgen flutamide. In isolated Sertoli and peritubular cells, testosterone pre-treatment suppressed the LPS-induced inflammatory response on TNF-α mRNA expression, while no effect was evident in testicular macrophages (TM). Conclusions Androgens can influence the immune system under normal conditions by the generation and functional differentiation of regulatory T cells and in testicular inflammation by direct effect on Sertoli and peritubular cells.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.