1
|
Sun F, Desevin K, Fu Y, Parameswaran S, Mayall J, Rinaldi V, Krietenstein N, Manukyan A, Yin Q, Galan C, Yang CH, Shindyapina AV, Gladyshev VN, Garber M, Schjenken JE, Rando OJ. A single cell atlas of the mouse seminal vesicle. G3 (BETHESDA, MD.) 2025; 15:jkaf045. [PMID: 40036847 PMCID: PMC12060236 DOI: 10.1093/g3journal/jkaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/16/2025] [Indexed: 03/06/2025]
Abstract
During mammalian reproduction, sperm are delivered to the female reproductive tract bathed in a complex medium known as seminal fluid, which plays key roles in signaling to the female reproductive tract and in nourishing sperm for their onwards journey. Along with minor contributions from the prostate and the epididymis, the majority of seminal fluid is produced by a somewhat understudied organ known as the seminal vesicle. Here, we report the first single-cell RNA-seq atlas of the mouse seminal vesicle, generated using tissues obtained from 23 mice of varying ages, exposed to a range of dietary challenges. We define the transcriptome of the secretory cells in this tissue, identifying a relatively homogeneous population of the epithelial cells which are responsible for producing the majority of seminal fluid. We also define the immune cell populations-including large populations of macrophages, dendritic cells, T cells, and NKT cells-which have the potential to play roles in producing the various immune mediators present in seminal plasma. Together, our data provide a resource for understanding the composition of an understudied reproductive tissue, with potential implications for paternal control of offspring development and metabolism.
Collapse
Affiliation(s)
- Fengyun Sun
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Kathleen Desevin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Yu Fu
- Department of Systems Biology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Shanmathi Parameswaran
- Hunter Medical Research Institute Infertility and Reproduction Research Program, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2305, Australia
| | - Jemma Mayall
- Immune Health Program, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Vera Rinaldi
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Nils Krietenstein
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Artür Manukyan
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Qiangzong Yin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Carolina Galan
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Chih-Hsiang Yang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Manuel Garber
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - John E Schjenken
- Hunter Medical Research Institute Infertility and Reproduction Research Program, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2305, Australia
| | - Oliver J Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| |
Collapse
|
2
|
Lawrence BM, O’Donnell L, Gannon AL, Skerrett-Byrne DA, Parameswaran S, Abbott I, Smith S, Handelsman DJ, Rebourcet D, Smith LB. Functional Analysis of HSD17B3-Deficient Male Mice Reveals Roles for HSD17B7 and HSD17B12 in Testosterone Biosynthesis. Endocrinology 2025; 166:bqaf078. [PMID: 40336300 PMCID: PMC12059215 DOI: 10.1210/endocr/bqaf078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Indexed: 05/09/2025]
Abstract
Historically, 17β-hydroxysteroid dehydrogenase type 3 (HSD17B3) was thought to be the key enzyme responsible for testicular testosterone production. In humans, loss-of-function mutations in HSD17B3 impair testosterone production during prenatal life leading to impaired development of androgen-dependent tissues in 46,XY individuals. However, male mice with HSD17B3 deficiency exhibit normal testicular testosterone concentrations, normal development of reproductive organs and are fertile, suggesting that mice express other hydroxysteroid dehydrogenase enzymes capable of testicular testosterone synthesis. This study aimed to investigate whether 17β-hydroxysteroid dehydrogenase type 12 (HSD17B12), which can convert androstenedione to testosterone in mice but not in humans, compensates for the lack of HSD17B3 in Hsd17b3 knockout (KO) mice. We used CRISPR/Cas9 to substitute the amino acid in mouse HSD17B12 that is responsible for its ability to convert androstenedione to testosterone with the amino acid of the human enzyme that prevents androstenedione being used as a substrate. When this Hsd17b12 mutation was introduced into Hsd17b3 KO mice, males exhibited normal reproductive tracts but reduced testicular testosterone production with a consequential reduction in seminal vesicle weight. This suggests HSD17B12 contributes toward testosterone production in the absence of HSD17B3, but other enzymes must also contribute. We therefore quantified other testicular hydroxysteroid dehydrogenases, finding that HSD17B7 mRNA and protein was markedly upregulated in Hsd17b3 KO testes. We confirmed that mouse, but not human, HSD17B7 can produce testosterone in vitro. We conclude that compared to humans, mice exhibit increased plasticity in testosterone production via hydroxysteroid dehydrogenase enzymes to support androgen action and male fertility.
Collapse
Affiliation(s)
- Ben M Lawrence
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Liza O’Donnell
- Office of the Deputy Vice-Chancellor (Research), Griffith University, Southport, Queensland 4222, Australia
- Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Anne-Louise Gannon
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales 2305, Australia
| | - David A Skerrett-Byrne
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales 2305, Australia
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, New South Wales 2308, Australia
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Shanmathi Parameswaran
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Imogen Abbott
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Sarah Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - David J Handelsman
- University of Sydney and Andrology Department, ANZAC Research Institute, Concord Hospital, Sydney, New South Wales 2139, Australia
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
- Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, Univ Rennes, Inserm, EHESP, Rennes 35065, France
| | - Lee B Smith
- Office of the Deputy Vice-Chancellor (Research), Griffith University, Southport, Queensland 4222, Australia
| |
Collapse
|
3
|
Perera TRW, de Ruijter‐Villani M, Gibb Z, Nixon B, Sheridan A, Stout TAE, Swegen A, Skerrett‐Byrne DA. Systemic Changes in Early Pregnancy in the Mare: An Integrated Proteomic Analysis of Blood Plasma, Histotroph, and Yolk Sac Fluid at Day 14 Post-Ovulation. Proteomics Clin Appl 2025; 19:e202400095. [PMID: 39912552 PMCID: PMC11895760 DOI: 10.1002/prca.202400095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE Embryo-maternal signaling during the establishment of pregnancy in horses remains one of the biggest mysteries in large animal physiology. Early pregnancy loss represents a major source of economic loss to the breeding industry. This study aimed to investigate the systemic changes associated with early pregnancy by mapping the proteome of blood plasma at 14 days in pregnant and non-pregnant mares. EXPERIMENTAL DESIGN Plasma proteomes were analysed in commercially bred pregnant (n = 17) and non-pregnant (n = 17) Thoroughbred mares at 14 days after ovulation, using high-resolution mass spectrometry. Day 14 histotroph and yolk sac fluid were also profiled and datasets were integrated through pathway analysis. RESULTS We identified 229 total protein IDs, with 12 increased and 10 decreased significantly in pregnant versus non-pregnant plasma. To gain functional insight, these data were aligned with proteomes of 14-day pregnant mare uterine fluid (n = 4; 1358 IDs) and conceptus fluid (soluble proteins within the yolk sac fluid; n = 4; 1152 IDs), and further interrogated using gene ontology databases and pathway analysis. CONCLUSIONS AND CLINICAL RELEVANCE These analyses identified consistent systemic changes in the mare's proteome that indicate a profound and specific immune response to early pregnancy, which appears to precede the systemic endocrine response to pregnancy. Integrated pathway analysis suggests that embryo-maternal interactions in early pregnancy may mimic elements of the virus-host interaction to modulate the maternal immune response. Transthyretin (TTR) and uteroglobin (SCGB1A1) were respectively down- and upregulated in plasma while also present in uterine fluid, and are proposed to be key proteins in early pregnancy establishment. These findings contribute significantly to our knowledge of early pregnancy in the mare and identify potential new avenues for developing clinical approaches to reduce early embryo loss.
Collapse
Affiliation(s)
- Tharangani R. W. Perera
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | | | - Zamira Gibb
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Brett Nixon
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Alecia Sheridan
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Tom A. E. Stout
- Department of Clinical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Aleona Swegen
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - David A. Skerrett‐Byrne
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
- Institute of Experimental GeneticsHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
4
|
Chan HY, Schjenken JE, Sharkey DJ, Robertson SA. Assessing Seminal Fluid Interaction with Female Reproductive Tract Tissues: In Vivo and In Vitro Models for Mouse. Methods Mol Biol 2025; 2897:649-662. [PMID: 40202667 DOI: 10.1007/978-1-0716-4406-5_44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
After intromission of seminal fluid at mating in mammals, the seminal fluid interacts with the lining of the female reproductive tract and elicits a female immune response. This response is triggered by signals in seminal fluid (carried by both the sperm and seminal plasma components) that bind receptors on female tract epithelial cells, causing a change in their transcriptional program to upregulate expression of pro-inflammatory cytokines and chemokines. The ensuing leukocyte recruitment and immune activation facilitates conception, embryo development, and progression of pregnancy. In this chapter, we report methods for evaluating effects of seminal fluid and its components (seminal plasma and sperm) on the female reproductive tract immune response in mice.
Collapse
Affiliation(s)
- Hon Yeung Chan
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - David J Sharkey
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Sarah A Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
5
|
Trigg N, Schjenken JE, Martin JH, Skerrett-Byrne DA, Smyth SP, Bernstein IR, Anderson AL, Stanger SJ, Simpson ENA, Tomar A, Teperino R, Conine CC, De Iuliis GN, Roman SD, Bromfield EG, Dun MD, Eamens AL, Nixon B. Subchronic elevation in ambient temperature drives alterations to the sperm epigenome and accelerates early embryonic development in mice. Proc Natl Acad Sci U S A 2024; 121:e2409790121. [PMID: 39527742 PMCID: PMC11588121 DOI: 10.1073/pnas.2409790121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Forecasted increases in the prevalence and severity of extreme weather events accompanying changes in climatic behavior pose potential risk to the reproductive capacity of humans and animals of ecological and agricultural significance. While several studies have revealed that heat stress induced by challenges such as testicular insulation can elicit a marked negative effect on the male reproductive system, and particularly the production of spermatozoa, less is known about the immediate impact on male reproductive function following subchronic whole-body exposure to elevated ambient temperature. To address this knowledge gap, we exposed unrestrained male mice to heat stress conditions that emulate a heat wave (daily cycle of 8 h at 35 °C followed by 16 h at 25 °C) for a period of 7 d. Neither the testes or epididymides of heat-exposed male mice exhibited evidence of gross histological change, and similarly, spermatozoa of exposed males retained their functionality and ability to support embryonic development. However, the embryos generated from heat-exposed spermatozoa experienced pronounced changes in gene expression linked to acceleration of early embryo development, aberrant blastocyst hatching, and increased fetal:placental weight ratio. Such changes were causally associated with an altered sperm small noncoding RNA (sncRNA) profile, such that these developmental phenotypes were recapitulated by microinjection of wild-type embryos sired by control spermatozoa with RNAs extracted from heat-exposed spermatozoa. Such data highlight that even relatively modest excursions in ambient temperature can affect male reproductive function and identify the sperm sncRNA profile as a particular point of vulnerability to this imposed environmental stress.
Collapse
Affiliation(s)
- Natalie Trigg
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- Department of Genetics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Department of Pediatrics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Center for Reproductive and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - John E. Schjenken
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Jacinta H. Martin
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - David A. Skerrett-Byrne
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Shannon P. Smyth
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- School of BioSciences Bio21 Molecular Sciences and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, VIC3010, Australia
| | - Ilana R. Bernstein
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Amanda L. Anderson
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Simone J. Stanger
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Ewan N. A. Simpson
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Archana Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Colin C. Conine
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Department of Pediatrics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Center for Reproductive and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Geoffry N. De Iuliis
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Shaun D. Roman
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- NSW Health Pathology, Newcastle, NSW2300, Australia
| | - Elizabeth G. Bromfield
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- School of BioSciences Bio21 Molecular Sciences and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, VIC3010, Australia
| | - Matthew D. Dun
- Cancer Signaling Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Andrew L. Eamens
- School of Health, University of the Sunshine Coast, Maroochydore, QLD4558, Australia
| | - Brett Nixon
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| |
Collapse
|
6
|
Skerrett-Byrne DA, Stanger SJ, Trigg NA, Anderson AL, Sipilä P, Bernstein IR, Lord T, Schjenken JE, Murray HC, Verrills NM, Dun MD, Pang TY, Nixon B. Phosphoproteomic analysis of the adaption of epididymal epithelial cells to corticosterone challenge. Andrology 2024; 12:1038-1057. [PMID: 38576152 DOI: 10.1111/andr.13636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND The epididymis has long been of interest owing to its role in promoting the functional maturation of the male germline. More recent evidence has also implicated the epididymis as an important sensory tissue responsible for remodeling of the sperm epigenome, both under physiological conditions and in response to diverse forms of environmental stress. Despite this knowledge, the intricacies of the molecular pathways involved in regulating the adaptation of epididymal tissue to paternal stressors remains to be fully resolved. OBJECTIVE The overall objective of this study was to investigate the direct impact of corticosterone challenge on a tractable epididymal epithelial cell line (i.e., mECap18 cells), in terms of driving adaptation of the cellular proteome and phosphoproteome signaling networks. MATERIALS AND METHODS The newly developed phosphoproteomic platform EasyPhos coupled with sequencing via an Orbitrap Exploris 480 mass spectrometer, was applied to survey global changes in the mECap18 cell (phospho)proteome resulting from sub-chronic (10-day) corticosterone challenge. RESULTS The imposed corticosterone exposure regimen elicited relatively subtle modifications of the global mECap18 proteome (i.e., only 73 out of 4171 [∼1.8%] proteins displayed altered abundance). By contrast, ∼15% of the mECap18 phosphoproteome was substantially altered following corticosterone challenge. In silico analysis of the corresponding parent proteins revealed an activation of pathways linked to DNA damage repair and oxidative stress responses as well as a reciprocal inhibition of pathways associated with organismal death. Corticosterone challenge also induced the phosphorylation of several proteins linked to the biogenesis of microRNAs. Accordingly, orthogonal validation strategies confirmed an increase in DNA damage, which was ameliorated upon selective kinase inhibition, and an altered abundance profile of a subset of microRNAs in corticosterone-treated cells. CONCLUSIONS Together, these data confirm that epididymal epithelial cells are reactive to corticosterone challenge, and that their response is tightly coupled to the opposing action of cellular kinases and phosphatases.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Petra Sipilä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| | - Terence Y Pang
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, New Lambton, NSW, Australia
| |
Collapse
|
7
|
Darmani H, Ali RRA. Long-term effect of exposure to triethylene glycol dimethacrylate (TEGDMA) on male mouse reproduction. Eur J Oral Sci 2024; 132:e12988. [PMID: 38664917 DOI: 10.1111/eos.12988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 05/16/2024]
Abstract
Our study investigated the impact on male mouse fertility and reproduction of long-term (14 weeks) exposure to triethylene glycol dimethacrylate (TEGDMA), a co-monomer of resin-based compounds, at doses of 0.01, 0.1, 1, and 10 ppm. Test and control mice were then paired with sexually mature untreated female mice and their fertility evaluated. Females paired with males exposed to all TEGDMA doses exhibited a significant decline in pregnancy rates, and significant increases in the total embryonic resorption-to-implantation ratio, except for males exposed to 0.01 ppm TEGDMA. Males in the highest dose group (10 ppm) showed significant increases in seminal vesicle and preputial gland weights. They also had significantly higher serum levels of luteinizing hormone (LH) and follicle stimulating hormone (FSH) than the controls, and the 0.01 ppm dosage group for FSH levels. TEGDMA exposure resulted in notable histopathological alterations in the testis, with detachment of germ cells and shedding of germinal epithelium into the tubule lumen. These results strongly indicate that TEGDMA exposure has detrimental consequences on the reproductive abilities and functions in male mice through disruption of the standard hormonal regulation of the reproductive system, leading to changes in spermatogenesis and ultimately leading to decreased fertility.
Collapse
Affiliation(s)
- Homa Darmani
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Razan Ramiz Abbas Ali
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
8
|
Murray HC, Miller K, Dun MD, Verrills NM. Pharmaco-phosphoproteomic analysis of cancer-associated KIT mutations D816V and V560G. Proteomics 2024; 24:e2300309. [PMID: 38334196 DOI: 10.1002/pmic.202300309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/24/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
The CD117 mast/stem cell growth factor receptor tyrosine kinase (KIT) is critical for haematopoiesis, melanogenesis and stem cell maintenance. KIT is commonly activated by mutation in cancers including acute myeloid leukaemia, melanoma and gastrointestinal stromal tumours (GISTs). The kinase and the juxtamembrane domains of KIT are mutation hotspots; with the kinase domain mutation D816V common in leukaemia and the juxtamembrane domain mutation V560G common in GISTs. Given the importance of mutant KIT signalling in cancer, we have conducted a proteomic and phosphoproteomic analysis of myeloid progenitor cells expressing D816V- and V560G-KIT mutants, using an FDCP1 isogenic cell line model. Proteomic analysis revealed increased abundance of proteases and growth signalling proteins in KIT-mutant cells compared to empty vector (EV) controls. Pathway analysis identified increased oxidative phosphorylation in D816V- and V560G-mutant KIT cells, which was targetable using the inhibitor IACS010759. Dysregulation of RNA metabolism and cytoskeleton/adhesion pathways was identified in both the proteome and phosphoproteome of KIT-mutant cells. Phosphoproteome analysis further revealed active kinases such as EGFR, ERK and PKC, which were targetable using pharmacological inhibitors. This study provides a pharmaco-phosphoproteomic profile of D816V- and V560G-mutant KIT cells, which reveals novel therapeutic strategies that may be applicable to a range of cancers.
Collapse
Affiliation(s)
- Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Kasey Miller
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
9
|
Sun F, Desevin K, Fu Y, Parameswaran S, Mayall J, Rinaldi V, Krietenstein N, Manukyan A, Yin Q, Galan C, Yang CH, Shindyapina AV, Gladyshev VN, Garber M, Schjenken JE, Rando OJ. A single cell atlas of the mouse seminal vesicle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588538. [PMID: 38645090 PMCID: PMC11030459 DOI: 10.1101/2024.04.08.588538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
During mammalian reproduction, sperm are delivered to the female reproductive tract bathed in a complex medium known as seminal fluid, which plays key roles in signaling to the female reproductive tract and in nourishing sperm for their onwards journey. Along with minor contributions from the prostate and the epididymis, the majority of seminal fluid is produced by a somewhat understudied organ known as the seminal vesicle. Here, we report the first single-cell RNA-seq atlas of the mouse seminal vesicle, generated using tissues obtained from 23 mice of varying ages, exposed to a range of dietary challenges. We define the transcriptome of the secretory cells in this tissue, identifying a relatively homogeneous population of the epithelial cells which are responsible for producing the majority of seminal fluid. We also define the immune cell populations - including large populations of macrophages, dendritic cells, T cells, and NKT cells - which have the potential to play roles in producing various immune mediators present in seminal plasma. Together, our data provide a resource for understanding the composition of an understudied reproductive tissue with potential implications for paternal control of offspring development and metabolism.
Collapse
Affiliation(s)
- Fengyun Sun
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kathleen Desevin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Yu Fu
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shanmathi Parameswaran
- Hunter Medical Research Institute Infertility and Reproduction Research Program, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia
| | - Jemma Mayall
- Priority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Vera Rinaldi
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nils Krietenstein
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Artur Manukyan
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Qiangzong Yin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Carolina Galan
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Chih-Hsiang Yang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anastasia V. Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Manuel Garber
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - John E. Schjenken
- Hunter Medical Research Institute Infertility and Reproduction Research Program, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia
| | - Oliver J. Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
10
|
Mulhall JE, Trigg NA, Bernstein IR, Anderson AL, Murray HC, Sipilä P, Lord T, Schjenken JE, Nixon B, Skerrett-Byrne DA. Immortalized mouse caput epididymal epithelial (mECap18) cell line recapitulates the in-vivo environment. Proteomics 2024; 24:e2300253. [PMID: 37759396 DOI: 10.1002/pmic.202300253] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/01/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Residing between the testes and the vas deferens, the epididymis is a highly convoluted tubule whose unique luminal microenvironment is crucial for the functional maturation of spermatozoa. This microenvironment is created by the combined secretory and resorptive activity of the lining epididymal epithelium, including the release of extracellular vesicles (epididymosomes), which encapsulate fertility modulating proteins and a myriad of small non-coding RNAs (sncRNAs) that are destined for delivery to recipient sperm cells. To enable investigation of this intercellular communication nexus, we have previously developed an immortalized mouse caput epididymal epithelial cell line (mECap18). Here, we describe the application of label-free mass spectrometry to characterize the mECap18 cell proteome and compare this to the proteome of native mouse caput epididymal epithelial cells. We report the identification of 5,313 mECap18 proteins, as many as 75.8% of which were also identified in caput epithelial cells wherein they mapped to broadly similar protein classification groupings. Furthermore, key pathways associated with protein synthesis (e.g., EIF2 signaling) and cellular protection in the male reproductive tract (e.g., sirtuin signaling) were enriched in both proteomes. This comparison supports the utility of the mECap18 cell line as a tractable in-vitro model for studying caput epididymal epithelial cell function.
Collapse
Affiliation(s)
- Jess E Mulhall
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Natalie A Trigg
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Departments of Genetics and Pediatrics - Penn Epigenetics Institute, Institute of Regenerative Medicine, and Center for Research on Reproduction and Women's Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, New South Wales, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Petra Sipilä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, and Turku Center for Disease Modeling, University of Turku, Turku, Varsinais-Suomi, Finland
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| |
Collapse
|
11
|
Chen Y, Roselli S, Panicker N, Brzozowski JS, Skerrett-Byrne DA, Murray HC, Verrills NM. Proteomic and phosphoproteomic characterisation of primary mouse embryonic fibroblasts. Proteomics 2024; 24:e2300267. [PMID: 37849217 DOI: 10.1002/pmic.202300267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
Fibroblasts are the most common cell type in stroma and function in the support and repair of most tissues. Mouse embryonic fibroblasts (MEFs) are amenable to isolation and rapid growth in culture. MEFs are therefore widely used as a standard model for functional characterisation of gene knockouts, and can also be used in co-cultures, commonly to support embryonic stem cell cultures. To facilitate their use as a research tool, we have performed a comprehensive proteomic and phosphoproteomic characterisation of wild-type primary MEFs from C57BL/6 mice. EIF2/4 and MTOR signalling pathways were abundant in both the proteome and phosphoproteome, along with extracellular matrix (ECM) and cytoskeleton associated pathways. Consistent with this, kinase enrichment analysis identified activation of P38A, P90RSK, P70S6K, and MTOR. Cell surface markers and matrisome proteins were also annotated. Data are available via ProteomeXchange with identifier PXD043244. This provides a comprehensive catalogue of the wild-type MEF proteome and phosphoproteome which can be utilised by the field to guide future work.
Collapse
Affiliation(s)
- Yanfang Chen
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute, University of Newcastle; and Precision Medicine Program, Callaghan, New South Wales, Australia
| | - Severine Roselli
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute, University of Newcastle; and Precision Medicine Program, Callaghan, New South Wales, Australia
| | - Nikita Panicker
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute, University of Newcastle; and Cancer Detection and Therapies Program, Callaghan, New South Wales, Australia
| | - Joshua S Brzozowski
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute, University of Newcastle; and Precision Medicine Program, Callaghan, New South Wales, Australia
| | - David A Skerrett-Byrne
- The Priority Research Centre for Reproductive Science, Hunter Medical Research Institute, The University of Newcastle; and the Infertility and Reproduction Research Program, Callaghan, New South Wales, Australia
| | - Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute, University of Newcastle; and Precision Medicine Program, Callaghan, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute, University of Newcastle; and Precision Medicine Program, Callaghan, New South Wales, Australia
| |
Collapse
|
12
|
Shen Q, Wu X, Chen J, He C, Wang Z, Zhou B, Zhang H. Immune Regulation of Seminal Plasma on the Endometrial Microenvironment: Physiological and Pathological Conditions. Int J Mol Sci 2023; 24:14639. [PMID: 37834087 PMCID: PMC10572377 DOI: 10.3390/ijms241914639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Seminal plasma (SP) accounts for more than 90% of semen volume. It induces inflammation, regulates immune tolerance, and facilitates embryonic development and implantation in the female reproductive tract. In the physiological state, SP promotes endometrial decidualization and causes changes in immune cells such as macrophages, natural killer cells, regulatory T cells, and dendritic cells. This leads to the secretion of cytokines and chemokines and also results in the alteration of miRNA profiles and the expression of genes related to endometrial tolerance and angiogenesis. Together, these changes modulate the endometrial immune microenvironment and contribute to implantation and pregnancy. However, in pathological situations, abnormal alterations in SP due to advanced age or poor diet in men can interfere with a woman's immune adaptation to pregnancy, negatively affecting embryo implantation and even the health of the offspring. Uterine pathologies such as endometriosis and endometritis can cause the endometrium to respond negatively to SP, which can further contribute to pathological progress and interfere with conception. The research on the mechanism of SP in the endometrium is conducive to the development of new targets for intervention to improve reproductive outcomes and may also provide new ideas for semen-assisted treatment of clinical infertility.
Collapse
Affiliation(s)
- Qiuzi Shen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.S.); (X.W.); (J.C.); (C.H.)
| | - Xiaoyu Wu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.S.); (X.W.); (J.C.); (C.H.)
| | - Jin Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.S.); (X.W.); (J.C.); (C.H.)
| | - Chao He
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.S.); (X.W.); (J.C.); (C.H.)
| | - Zehao Wang
- School of Management, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Boyan Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.S.); (X.W.); (J.C.); (C.H.)
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.S.); (X.W.); (J.C.); (C.H.)
| |
Collapse
|
13
|
Wu SL, Ju JQ, Ji YM, Zhang HL, Zou YJ, Sun SC. Exposure to acrylamide induces zygotic genome activation defects of mouse embryos. Food Chem Toxicol 2023; 175:113753. [PMID: 36997053 DOI: 10.1016/j.fct.2023.113753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Acrylamide (ACR) is an important chemical raw material for wastewater treatment, paper industry and textile industry, which is widely exposed from occupational, environmental and dietary situation. ACR has neurotoxicity, genotoxicity, potential carcinogenicity and reproductive toxicity. Recent study indicates that ACR affected oocyte maturation quality. In the present study, we reported the effects of ACR exposure on zygotic genome activation (ZGA) in embryos and its related mechanism. Our results showed that ACR treatment caused 2-cell arrest in mouse embryos, indicating the failure of ZGA, which was confirmed by decreased global transcription levels and aberrant expression of ZGA-related and maternal factors. We found that histone modifications such as H3K9me3, H3K27me3 and H3K27ac levels were altered, and this might be due to the occurrence of DNA damage, showing with positive γ-H2A.X signal. Moreover, mitochondrial dysfunction and high levels of ROS were detected in ACR treated embryos, indicating that ACR induced oxidative stress, and this might further cause abnormal distribution of endoplasmic reticulum, Golgi apparatus and lysosomes. In conclusion, our results indicated that ACR exposure disrupted ZGA by inducing mitochondria-based oxidative stress, which further caused DNA damage, aberrant histone modifications and organelles in mouse embryos.
Collapse
Affiliation(s)
- Si-Le Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yi-Ming Ji
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hao-Lin Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yuan-Jing Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
14
|
Murray HC, Miller K, Brzozowski JS, Kahl RGS, Smith ND, Humphrey SJ, Dun MD, Verrills NM. Synergistic Targeting of DNA-PK and KIT Signaling Pathways in KIT Mutant Acute Myeloid Leukemia. Mol Cell Proteomics 2023; 22:100503. [PMID: 36682716 PMCID: PMC9986649 DOI: 10.1016/j.mcpro.2023.100503] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 12/19/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common and aggressive form of acute leukemia, with a 5-year survival rate of just 24%. Over a third of all AML patients harbor activating mutations in kinases, such as the receptor tyrosine kinases FLT3 (receptor-type tyrosine-protein kinase FLT3) and KIT (mast/stem cell growth factor receptor kit). FLT3 and KIT mutations are associated with poor clinical outcomes and lower remission rates in response to standard-of-care chemotherapy. We have recently identified that the core kinase of the non-homologous end joining DNA repair pathway, DNA-PK (DNA-dependent protein kinase), is activated downstream of FLT3; and targeting DNA-PK sensitized FLT3-mutant AML cells to standard-of-care therapies. Herein, we investigated DNA-PK as a possible therapeutic vulnerability in KIT mutant AML, using isogenic FDC-P1 mouse myeloid progenitor cell lines transduced with oncogenic mutant KIT (V560G and D816V) or vector control. Targeted quantitative phosphoproteomic profiling identified phosphorylation of DNA-PK in the T2599/T2605/S2608/S2610 cluster in KIT mutant cells, indicative of DNA-PK activation. Accordingly, proliferation assays revealed that KIT mutant FDC-P1 cells were more sensitive to the DNA-PK inhibitors M3814 or NU7441, compared with empty vector controls. DNA-PK inhibition combined with inhibition of KIT signaling using the kinase inhibitors dasatinib or ibrutinib, or the protein phosphatase 2A activators FTY720 or AAL(S), led to synergistic cell death. Global phosphoproteomic analysis of KIT-D816V cells revealed that dasatinib and M3814 single-agent treatments inhibited extracellular signal-regulated kinase and AKT (RAC-alpha serine/threonine-protein kinase)/MTOR (serine/threonine-protein kinase mTOR) activity, with greater inhibition of both pathways when used in combination. Combined dasatinib and M3814 treatment also synergistically inhibited phosphorylation of the transcriptional regulators MYC and MYB. This study provides insight into the oncogenic pathways regulated by DNA-PK beyond its canonical role in DNA repair and demonstrates that DNA-PK is a promising therapeutic target for KIT mutant cancers.
Collapse
Affiliation(s)
- Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Kasey Miller
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Joshua S Brzozowski
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Richard G S Kahl
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Nathan D Smith
- Analytical and Biomolecular Research Facility, Advanced Mass Spectrometry Unit, University of Newcastle, Callaghan, New South Wales, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, and The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia.
| |
Collapse
|
15
|
Nixon B, Schjenken JE, Burke ND, Skerrett-Byrne DA, Hart HM, De Iuliis GN, Martin JH, Lord T, Bromfield EG. New horizons in human sperm selection for assisted reproduction. Front Endocrinol (Lausanne) 2023; 14:1145533. [PMID: 36909306 PMCID: PMC9992892 DOI: 10.3389/fendo.2023.1145533] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Male infertility is a commonly encountered pathology that is estimated to be a contributory factor in approximately 50% of couples seeking recourse to assisted reproductive technologies. Upon clinical presentation, such males are commonly subjected to conventional diagnostic andrological practices that rely on descriptive criteria to define their fertility based on the number of morphologically normal, motile spermatozoa encountered within their ejaculate. Despite the virtual ubiquitous adoption of such diagnostic practices, they are not without their limitations and accordingly, there is now increasing awareness of the importance of assessing sperm quality in order to more accurately predict a male's fertility status. This realization raises the important question of which characteristics signify a high-quality, fertilization competent sperm cell. In this review, we reflect on recent advances in our mechanistic understanding of sperm biology and function, which are contributing to a growing armory of innovative approaches to diagnose and treat male infertility. In particular we review progress toward the implementation of precision medicine; the robust clinical adoption of which in the setting of fertility, currently lags well behind that of other fields of medicine. Despite this, research shows that the application of advanced technology platforms such as whole exome sequencing and proteomic analyses hold considerable promise in optimizing outcomes for the management of male infertility by uncovering and expanding our inventory of candidate infertility biomarkers, as well as those associated with recurrent pregnancy loss. Similarly, the development of advanced imaging technologies in tandem with machine learning artificial intelligence are poised to disrupt the fertility care paradigm by advancing our understanding of the molecular and biological causes of infertility to provide novel avenues for future diagnostics and treatments.
Collapse
Affiliation(s)
- Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- *Correspondence: Brett Nixon,
| | - John E. Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nathan D. Burke
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - David A. Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Hanah M. Hart
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Geoffry N. De Iuliis
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jacinta H. Martin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Elizabeth G. Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
16
|
Skerrett-Byrne DA, Anderson AL, Bromfield EG, Bernstein IR, Mulhall JE, Schjenken JE, Dun MD, Humphrey SJ, Nixon B. Global profiling of the proteomic changes associated with the post-testicular maturation of mouse spermatozoa. Cell Rep 2022; 41:111655. [DOI: 10.1016/j.celrep.2022.111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
|
17
|
Zhang M, Chiozzi RZ, Skerrett-Byrne DA, Veenendaal T, Klumperman J, Heck AJR, Nixon B, Helms JB, Gadella BM, Bromfield EG. High Resolution Proteomic Analysis of Subcellular Fractionated Boar Spermatozoa Provides Comprehensive Insights Into Perinuclear Theca-Residing Proteins. Front Cell Dev Biol 2022; 10:836208. [PMID: 35252197 PMCID: PMC8894813 DOI: 10.3389/fcell.2022.836208] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
The perinuclear theca (PT) is a highly condensed, largely insoluble protein structure that surrounds the nucleus of eutherian spermatozoa. Recent reports have indicated that the PT unexpectedly houses several somatic proteins, such as core histones, which may be important post-fertilization during re-modelling of the male pronucleus, yet little is known regarding the overall proteomic composition of the PT. Here, we report the first in depth, label-free proteomic characterization of the PT of boar spermatozoa following the implementation of a long-established subcellular fractionation protocol designed to increase the detection of low abundance proteins. A total of 1,802 proteins were identified, a result that represents unparalleled depth of coverage for the boar sperm proteome and exceeds the entire annotated proteome of the Sus scrofa species so far. In the PT structure itself, we identified 813 proteins and confirmed the presence of previously characterized PT proteins including the core histones H2A, H2B, H3 and H4, as well as Ras-related protein Rab-2A (RAB2A) and Rab-2B (RAB2B) amongst other RAB proteins. In addition to these previously characterized PT proteins, our data revealed that the PT is replete in proteins critical for sperm-egg fusion and egg activation, including: Izumo family members 1–4 (IZUMO1-4) and phosphoinositide specific phospholipase ζ (PLCZ1). Through Ingenuity Pathway Analysis, we found surprising enrichment of endoplasmic reticulum (ER) proteins and the ER-stress response in the PT. This is particularly intriguing as it is currently held that the ER structure is lost during testicular sperm maturation. Using the String and Cytoscape tools to visualize protein-protein interactions revealed an intricate network of PT protein complexes, including numerous proteasome subunits. Collectively, these data suggest that the PT may be a unique site of cellular homeostasis that houses an abundance of protein degradation machinery. This fits with previous observations that the PT structure dissociates first within the oocyte post-fertilization. It remains to be explored whether proteasome subunits within the PT actively assist in the protein degradation of paternal cell structures post-fertilization and how aberrations in PT protein content may delay embryonic development.
Collapse
Affiliation(s)
- Min Zhang
- Department of Biomolecular Health Sciences and Department of Farm and Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Netherlands Proteomics Centre, Utrecht, Netherlands
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - David A. Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Tineke Veenendaal
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Netherlands Proteomics Centre, Utrecht, Netherlands
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - J. Bernd Helms
- Department of Biomolecular Health Sciences and Department of Farm and Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Bart M. Gadella
- Department of Biomolecular Health Sciences and Department of Farm and Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Bart M. Gadella,
| | - Elizabeth G. Bromfield
- Department of Biomolecular Health Sciences and Department of Farm and Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
18
|
Griffin RA, Swegen A, Baker MA, Ogle RA, Smith N, Aitken RJ, Skerrett-Byrne DA, Fair S, Gibb Z. Proteomic analysis of spermatozoa reveals caseins play a pivotal role in preventing short-term periods of subfertility in stallions. Biol Reprod 2022; 106:741-755. [DOI: 10.1093/biolre/ioab225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/29/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
Stallions experience transient fluctuations in fertility throughout the breeding season. Considering pregnancy diagnoses cannot be ascertained until ~14 days post-breeding, the timely detection of decreases in stallion fertility would enhance industry economic and welfare outcomes. Therefore, this study aimed to identify the proteomic signatures reflective of short-term fertility fluctuations, and to determine the biological mechanisms governing such differences. Using LC–MS/MS, we compared the proteomic profile of semen samples collected from commercially “fertile” stallions, during high- and low-fertility periods. A total of 1702 proteins were identified, of which, 38 showed a significant change in abundance (p ≤ 0.05). Assessment of intra- and inter-stallion variability revealed that caseins (namely κ-, α-S1-, and α-S2-casein), were significantly more abundant during “high-fertility” periods, while several epididymal, and seminal plasma proteins (chiefly, epididymal sperm binding protein 1 [ELSPbP1], horse seminal plasma protein 1 [HSP-1] and clusterin), were significantly more abundant during “low-fertility” periods. We hypothesised that an increased abundance of caseins offers greater protection from potentially harmful seminal plasma proteins, thereby preserving cell functionality and fertility. In vitro exposure of spermatozoa to casein resulted in decreased levels of lipid scrambling (Merocyanine 540), higher abundance of sperm-bound caseins (α-S1-, α-S2-, and κ-casein), and lower abundance of sperm-bound HSP-1 (p ≤ 0.05). This study demonstrates key pathways governing short-term fertility fluctuations in the stallion, thereby providing a platform to develop robust, fertility assessment strategies into the future.
Collapse
Affiliation(s)
- Róisín Ann Griffin
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - Aleona Swegen
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Mark A Baker
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - Rachel Ann Ogle
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - Nathan Smith
- Analytical and Biomedical Research Facility, Research Division, University of Newcastle, Callaghan, New South Wales, Australia
| | - Robert John Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| | - David Anthony Skerrett-Byrne
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New South Wales, Australia
| | - Sean Fair
- Laboratory of Animal Reproduction, Department of Biological Sciences, Biomaterials Research Cluster, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Zamira Gibb
- Priority Research Centre for Reproductive Science, University of Newcastle, New South Wales, Australia
| |
Collapse
|
19
|
Smyth SP, Nixon B, Anderson AL, Murray HC, Martin JH, MacDougall LA, Robertson SA, Skerrett-Byrne DA, Schjenken JE. Elucidation of the protein composition of mouse seminal vesicle fluid. Proteomics 2022; 22:e2100227. [PMID: 35014747 DOI: 10.1002/pmic.202100227] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023]
Abstract
The seminal vesicles are male accessory sex glands that contribute the major portion of the seminal plasma in which mammalian spermatozoa are bathed during ejaculation. In addition to conveying sperm through the ejaculatory duct, seminal vesicle secretions support sperm survival after ejaculation, and influence the female reproductive tract to promote receptivity to pregnancy. Analysis of seminal vesicle fluid (SVF) composition by proteomics has proven challenging, due to its highly biased protein signature with a small subset of dominant proteins and the difficulty of solubilizing this viscous fluid. As such, publicly available proteomic datasets identify only 85 SVF proteins in total. To address this limitation, we report a new preparative methodology involving sequential solubilization of mouse SVF in guanidine hydrochloride, acetone precipitation, and analysis by label-free mass spectrometry. Using this strategy, we identified 126 SVF proteins, including 83 previously undetected in SVF. Members of the seminal vesicle secretory protein family were the most abundant, accounting for 79% of all peptide spectrum matches. Functional analysis identified inflammation and formation of the vaginal plug as the two most prominent biological processes. Other notable processes included modulation of sperm function and regulation of the female reproductive tract immune environment. Together, these findings provide a robust methodological framework for future SVF studies and identify novel proteins with potential to influence both male and female reproductive physiology.
Collapse
Affiliation(s)
- Shannon P Smyth
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Brett Nixon
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Amanda L Anderson
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia.,Precision Medicine Research Program, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Jacinta H Martin
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Lily A MacDougall
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - David A Skerrett-Byrne
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - John E Schjenken
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| |
Collapse
|
20
|
Simmons LW, Ng SH, Lovegrove M. Condition‐dependent seminal fluid gene expression and intergenerational paternal effects on ejaculate quality. Funct Ecol 2021. [DOI: 10.1111/1365-2435.13987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Leigh W. Simmons
- Centre for Evolutionary Biology School of Biological Sciences The University of Western Australia Crawley WA Australia
| | - Soon Hwee Ng
- Centre for Evolutionary Biology School of Biological Sciences The University of Western Australia Crawley WA Australia
| | - Maxine Lovegrove
- Centre for Evolutionary Biology School of Biological Sciences The University of Western Australia Crawley WA Australia
| |
Collapse
|
21
|
Skerrett-Byrne DA, Nixon B, Bromfield EG, Breen J, Trigg NA, Stanger SJ, Bernstein IR, Anderson AL, Lord T, Aitken RJ, Roman SD, Robertson SA, Schjenken JE. Transcriptomic analysis of the seminal vesicle response to the reproductive toxicant acrylamide. BMC Genomics 2021; 22:728. [PMID: 34625024 PMCID: PMC8499523 DOI: 10.1186/s12864-021-07951-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The seminal vesicles synthesise bioactive factors that support gamete function, modulate the female reproductive tract to promote implantation, and influence developmental programming of offspring phenotype. Despite the significance of the seminal vesicles in reproduction, their biology remains poorly defined. Here, to advance understanding of seminal vesicle biology, we analyse the mouse seminal vesicle transcriptome under normal physiological conditions and in response to acute exposure to the reproductive toxicant acrylamide. Mice were administered acrylamide (25 mg/kg bw/day) or vehicle control daily for five consecutive days prior to collecting seminal vesicle tissue 72 h following the final injection. RESULTS A total of 15,304 genes were identified in the seminal vesicles with those encoding secreted proteins amongst the most abundant. In addition to reproductive hormone pathways, functional annotation of the seminal vesicle transcriptome identified cell proliferation, protein synthesis, and cellular death and survival pathways as prominent biological processes. Administration of acrylamide elicited 70 differentially regulated (fold-change ≥1.5 or ≤ 0.67) genes, several of which were orthogonally validated using quantitative PCR. Pathways that initiate gene and protein synthesis to promote cellular survival were prominent amongst the dysregulated pathways. Inflammation was also a key transcriptomic response to acrylamide, with the cytokine, Colony stimulating factor 2 (Csf2) identified as a top-ranked upstream driver and inflammatory mediator associated with recovery of homeostasis. Early growth response (Egr1), C-C motif chemokine ligand 8 (Ccl8), and Collagen, type V, alpha 1 (Col5a1) were also identified amongst the dysregulated genes. Additionally, acrylamide treatment led to subtle changes in the expression of genes that encode proteins secreted by the seminal vesicle, including the complement regulator, Complement factor b (Cfb). CONCLUSIONS These data add to emerging evidence demonstrating that the seminal vesicles, like other male reproductive tract tissues, are sensitive to environmental insults, and respond in a manner with potential to exert impact on fetal development and later offspring health.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - James Breen
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,South Australian Genomics Centre (SAGC), South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.,Computational & Systems Biology Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia. .,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
22
|
Schjenken JE, Moldenhauer LM, Sharkey DJ, Chan HY, Chin PY, Fullston T, McPherson NO, Robertson SA. High-fat Diet Alters Male Seminal Plasma Composition to Impair Female Immune Adaptation for Pregnancy in Mice. Endocrinology 2021; 162:6309474. [PMID: 34170298 DOI: 10.1210/endocr/bqab123] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Indexed: 12/18/2022]
Abstract
Paternal experiences and exposures before conception can influence fetal development and offspring phenotype. The composition of seminal plasma contributes to paternal programming effects through modulating the female reproductive tract immune response after mating. To investigate whether paternal obesity affects seminal plasma immune-regulatory activity, C57Bl/6 male mice were fed an obesogenic high-fat diet (HFD) or control diet (CD) for 14 weeks. Although HFD consumption caused only minor changes to parameters of sperm quality, the volume of seminal vesicle fluid secretions was increased by 65%, and the concentrations and total content of immune-regulatory TGF-β isoforms were decreased by 75% to 80% and 43% to 55%, respectively. Mating with BALB/c females revealed differences in the strength and properties of the postmating immune response elicited. Transcriptional analysis showed >300 inflammatory genes were similarly regulated in the uterine endometrium by mating independently of paternal diet, and 13 were dysregulated by HFD-fed compared with CD-fed males. Seminal vesicle fluid factors reduced in HFD-fed males, including TGF-β1, IL-10, and TNF, were among the predicted upstream regulators of differentially regulated genes. Additionally, the T-cell response induced by mating with CD-fed males was blunted after mating with HFD-fed males, with 27% fewer CD4+ T cells, 26% fewer FOXP3+CD4+ regulatory T cells (Treg) cells, and 19% fewer CTLA4+ Treg cells, particularly within the NRP1+ thymic Treg cell population. These findings demonstrate that an obesogenic HFD alters the composition of seminal vesicle fluid and impairs seminal plasma capacity to elicit a favorable pro-tolerogenic immune response in females at conception.
Collapse
Affiliation(s)
- John E Schjenken
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Lachlan M Moldenhauer
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - David J Sharkey
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Hon Y Chan
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Peck Y Chin
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Tod Fullston
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
- Repromed, Dulwich, Adelaide, South Australia, 5065, Australia
| | - Nicole O McPherson
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
- Repromed, Dulwich, Adelaide, South Australia, 5065, Australia
- Freemasons Centre for Men's Health, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Sarah A Robertson
- The Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, 5005, Australia
- Monash IVF Group, Richmond, Victoria, 3121, Australia
| |
Collapse
|
23
|
Skerrett-Byrne DA, Anderson AL, Hulse L, Wass C, Dun MD, Bromfield EG, De Iuliis GN, Pyne M, Nicolson V, Johnston SD, Nixon B. Proteomic analysis of koala (phascolarctos cinereus) spermatozoa and prostatic bodies. Proteomics 2021; 21:e2100067. [PMID: 34411425 DOI: 10.1002/pmic.202100067] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/28/2021] [Accepted: 08/16/2021] [Indexed: 12/23/2022]
Abstract
The aims of this study were to investigate the proteome of koala spermatozoa and that of the prostatic bodies with which they interact during ejaculation. For this purpose, spermatozoa and prostatic bodies were fractionated from the semen of four male koalas and analysed by HPLC MS/MS. This strategy identified 744 sperm and 1297 prostatic body proteins, which were subsequently attributed to 482 and 776 unique gene products, respectively. Gene ontology curation of the sperm proteome revealed an abundance of proteins mapping to the canonical sirtuin and 14-3-3 signalling pathways. By contrast, protein ubiquitination and unfolded protein response pathways dominated the equivalent analysis of proteins uniquely identified in prostatic bodies. Koala sperm proteins featured an enrichment of those mapping to the functional categories of cellular compromise/inflammatory response, whilst those of the prostatic body revealed an over-representation of molecular chaperone and stress-related proteins. Cross-species comparisons demonstrated that the koala sperm proteome displays greater conservation with that of eutherians (human; 93%) as opposed to reptile (crocodile; 39%) and avian (rooster; 27%) spermatozoa. Together, this work contributes to our overall understanding of the core sperm proteome and has identified biomarkers that may contribute to the exceptional longevity of koala spermatozoa during ex vivo storage.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Lyndal Hulse
- School of Agriculture and Food Sciences, The University of Queensland, Gatton, Queensland, Australia
| | - Caillin Wass
- School of Agriculture and Food Sciences, The University of Queensland, Gatton, Queensland, Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Cancer Research Innovation and Translation, Hunter Medical Research Institute, Lambton, New South Wales, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Geoffry N De Iuliis
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Michael Pyne
- Currumbin Wildlife Sanctuary, Currumbin, Queensland, Australia
| | - Vere Nicolson
- Dreamworld, Dreamworld Parkway, Coomera, Queensland, Australia
| | - Stephen D Johnston
- School of Agriculture and Food Sciences, The University of Queensland, Gatton, Queensland, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
24
|
Skerrett-Byrne DA, Bromfield EG, Murray HC, Jamaluddin MFB, Jarnicki AG, Fricker M, Essilfie AT, Jones B, Haw TJ, Hampsey D, Anderson AL, Nixon B, Scott RJ, Wark PAB, Dun MD, Hansbro PM. Time-resolved proteomic profiling of cigarette smoke-induced experimental chronic obstructive pulmonary disease. Respirology 2021; 26:960-973. [PMID: 34224176 DOI: 10.1111/resp.14111] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/01/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Chronic obstructive pulmonary disease (COPD) is the third leading cause of illness and death worldwide. Current treatments aim to control symptoms with none able to reverse disease or stop its progression. We explored the major molecular changes in COPD pathogenesis. METHODS We employed quantitative label-based proteomics to map the changes in the lung tissue proteome of cigarette smoke-induced experimental COPD that is induced over 8 weeks and progresses over 12 weeks. RESULTS Quantification of 7324 proteins enabled the tracking of changes to the proteome. Alterations in protein expression profiles occurred in the induction phase, with 18 and 16 protein changes at 4- and 6-week time points, compared to age-matched controls, respectively. Strikingly, 269 proteins had altered expression after 8 weeks when the hallmark pathological features of human COPD emerge, but this dropped to 27 changes at 12 weeks with disease progression. Differentially expressed proteins were validated using other mouse and human COPD bronchial biopsy samples. Major changes in RNA biosynthesis (heterogeneous nuclear ribonucleoproteins C1/C2 [HNRNPC] and RNA-binding protein Musashi homologue 2 [MSI2]) and modulators of inflammatory responses (S100A1) were notable. Mitochondrial dysfunction and changes in oxidative stress proteins also occurred. CONCLUSION We provide a detailed proteomic profile, identifying proteins associated with the pathogenesis and disease progression of COPD establishing a platform to develop effective new treatment strategies.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Elizabeth G Bromfield
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia.,Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Heather C Murray
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - M Fairuz B Jamaluddin
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Ama T Essilfie
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Bernadette Jones
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Tatt J Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Daniel Hampsey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Amanda L Anderson
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Brett Nixon
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Rodney J Scott
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia
| | - Matthew D Dun
- University of Newcastle, Callaghan, New South Wales, Australia.,Cancer Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,University of Newcastle, Callaghan, New South Wales, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|