51
|
Papadopoulos D, Shihan M, Scheiner-Bobis G. Physiological implications of DHEAS-induced non-classical steroid hormone signaling. J Steroid Biochem Mol Biol 2018; 179:73-78. [PMID: 29017935 DOI: 10.1016/j.jsbmb.2017.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 09/28/2017] [Accepted: 10/03/2017] [Indexed: 01/12/2023]
Abstract
In the spermatogenic cell line GC-2, dehydroepiandrosterone sulfate (DHEAS), activates the Src/Ras/c-Raf/Erk1/2/CREB(ATF-1) signaling cascade. Since DHEAS is present in the gonads, and since spermatogenesis and maturation of spermatogonia to haploid spermatozoa requires activation of Erk1/2, the triggering of these signaling events by DHEAS might have physiological relevance. In the Sertoli cell line TM4, DHEAS-induces activation of Erk1/2, CREB, and ATF-1, stimulates expression of claudin-3 and claudin-5 and augments transepithelial resistance, indicating the formation of tight junctions between adjacent Sertoli cells. Thus, by influencing the formation and dynamics of tight junctions at the blood-testis barrier, which protects germ cells from cells of the immune system, DHEAS might play a crucial role in the regulation and maintenance of male fertility. In bEnd.3 brain-derived endothelial cells, DHEAS stimulates the expression of zonula occludens-1 and claudin-3 and promotes tight junction formation between neighboring cells, which at the blood-brain barrier protects the brain from harmful factors and cells. If DHEAS supports the integrity of the blood-brain barrier also in vivo, the current findings might lead to new strategies for the prevention or treatment of neurological disorders associated with barrier defects.
Collapse
Affiliation(s)
- Dimitrios Papadopoulos
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Germany
| | - Mazen Shihan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Germany.
| |
Collapse
|
52
|
Micati DJ, Hime GR, McLaughlin EA, Abud HE, Loveland KL. Differential expression profiles of conserved Snail transcription factors in the mouse testis. Andrology 2018; 6:362-373. [PMID: 29381885 DOI: 10.1111/andr.12465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/29/2017] [Accepted: 12/15/2017] [Indexed: 01/11/2023]
Abstract
Snail transcription factors are key regulators of cellular transitions during embryonic development and tumorigenesis. The closely related SNAI1 and SNAI2 proteins induce epithelial-mesenchymal transitions (EMTs), acting predominantly as transcriptional repressors, while the functions of SNAI3 are unknown. An initial examination of Snai2-deficient mice provided evidence of deficient spermatogenesis. To address the hypothesis that Snail proteins are important for male fertility, this study provides the first comprehensive cellular expression profiles of all three mammalian Snail genes in the post-natal mouse testis. To evaluate Snail transcript expression profiles, droplet digital (dd) PCR and in situ hybridization were employed. Snai1, 2 and 3 transcripts are readily detected at 7, 14, 28 days post-partum (dpp) and 7 weeks (adult). Unique cellular expression was demonstrated for each by in situ hybridization and immunohistochemistry using Western blot-validated antibodies. SNAI1 and SNAI2 are in the nucleus of the most mature germ cell types at post-natal ages 10, 15 and 26. SNAI3 is only detected from 15 dpp onwards and is localized in the Sertoli cell cytoplasm. In the adult testis, Snai1 and Snai2 transcripts are detected in spermatogonia and spermatocytes, while Snai3 is in both germ and Sertoli cells. SNAI1 protein is evident in nuclei of spermatogonia, spermatocytes, round spermatids and elongated spermatids (Stages IX-XII). SNAI2 is present in the nuclei of spermatogonia and spermatocytes, with a faint signal detected in round spermatids. SNAI3 was detected only in Sertoli cell cytoplasm, as in juvenile testes. Additionally, colocalization of SNAI1 and SNAI2 with previously identified key binding partners, LSD1 and PRC2 complex components, provides strong evidence that these important functional interactions are conserved during spermatogenesis to control gene activity. These distinct expression profiles suggest that each Snail family member has unique functions during spermatogenesis.
Collapse
Affiliation(s)
- D J Micati
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.,Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - G R Hime
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia
| | - E A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - H E Abud
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - K L Loveland
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.,Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
53
|
Bulldan A, Malviya VN, Upmanyu N, Konrad L, Scheiner-Bobis G. Testosterone/bicalutamide antagonism at the predicted extracellular androgen binding site of ZIP9. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2402-2414. [PMID: 28943399 DOI: 10.1016/j.bbamcr.2017.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 10/18/2022]
Abstract
ZIP9 is a Zn2+ transporter, testosterone receptor, and mediator of signaling events through G-proteins. Despite these pivotal properties, however, its physiological and pathophysiological significance has not yet been comprehensively addressed. Using a cell line that lacks the classical androgen receptor we show that ZIP9-mediated phosphorylation of Erk1/2, CREB, or ATF-1 and expression of claudin-5 and zonula occludens-1 by testosterone can be completely antagonized by bicalutamide (Casodex), an anti-androgen of significant clinical impact. Computational modeling and docking experiments with ZIP9 reveal typical characteristics of ZIP transporters and an extracellular binding site for testosterone capable of accommodating bicalutamide. The presence of this site is verified by our demonstration that the membrane-impermeable testosterone analogue T-BSA-FITC labels the membrane only when ZIP9 is expressed and that this labeling is completely prevented by bicalutamide. The study connects structural features of ZIP9 to its functions and indicates a possible relevance of ZIP9 as a pharmacological target.
Collapse
Affiliation(s)
- Ahmed Bulldan
- Institute for Veterinary-Physiology and -Biochemistry, School of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | | | - Neha Upmanyu
- Institute for Veterinary-Physiology and -Biochemistry, School of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany
| | - Lutz Konrad
- Department of Obstetrics and Gynecology, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institute for Veterinary-Physiology and -Biochemistry, School of Veterinary Medicine, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
54
|
Geyer J, Bakhaus K, Bernhardt R, Blaschka C, Dezhkam Y, Fietz D, Grosser G, Hartmann K, Hartmann MF, Neunzig J, Papadopoulos D, Sánchez-Guijo A, Scheiner-Bobis G, Schuler G, Shihan M, Wrenzycki C, Wudy SA, Bergmann M. The role of sulfated steroid hormones in reproductive processes. J Steroid Biochem Mol Biol 2017; 172:207-221. [PMID: 27392637 DOI: 10.1016/j.jsbmb.2016.07.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
Sulfated steroid hormones, such as dehydroepiandrosterone sulfate or estrone-3-sulfate, have long been regarded as inactive metabolites as they cannot activate classical steroid receptors. Some of them are present in the blood circulation at quite high concentrations, but generally sulfated steroids exhibit low membrane permeation due to their hydrophilic properties. However, sulfated steroid hormones can actively be imported into specific target cells via uptake carriers, such as the sodium-dependent organic anion transporter SOAT, and, after hydrolysis by the steroid sulfatase (so-called sulfatase pathway), contribute to the overall regulation of steroid responsive organs. To investigate the biological significance of sulfated steroid hormones for reproductive processes in humans and animals, the research group "Sulfated Steroids in Reproduction" was established by the German Research Foundation DFG (FOR1369). Projects of this group deal with transport of sulfated steroids, sulfation of free steroids, desulfation by the steroid sulfatase, effects of sulfated steroids on steroid biosynthesis and membrane receptors as well as MS-based profiling of sulfated steroids in biological samples. This review and concept paper presents key findings from all these projects and provides a broad overview over the current research on sulfated steroid hormones in the field of reproduction.
Collapse
Affiliation(s)
- Joachim Geyer
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany.
| | - Katharina Bakhaus
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Rita Bernhardt
- Institute of Biochemistry, Saarland University, Saarbrücken, Germany
| | - Carina Blaschka
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Yaser Dezhkam
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Gary Grosser
- Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Katja Hartmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Michaela F Hartmann
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Jens Neunzig
- Institute of Biochemistry, Saarland University, Saarbrücken, Germany
| | - Dimitrios Papadopoulos
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Alberto Sánchez-Guijo
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Gerhard Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Mazen Shihan
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University, Giessen, Germany
| | - Christine Wrenzycki
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus Liebig University, Giessen, Germany
| | - Stefan A Wudy
- Steroid Research & Mass Spectrometry Unit, Laboratory for Translational Hormone Analytics, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Martin Bergmann
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
55
|
Elayapillai SP, Teekaraman D, Paulraj RS, Jagadeesan A. Ameliorative effect of α-tocopherol on polychlorinated biphenyl (PCBs) induced testicular Sertoli cell dysfunction in F 1 prepuberal rats. ACTA ACUST UNITED AC 2017; 69:681-694. [PMID: 28739394 DOI: 10.1016/j.etp.2017.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/23/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022]
Abstract
The study was conducted to investigate the protective role of α-tocopherol against polychlorinated biphenyls (PCBs) induced effect in Sertoli cell function of F1 prepuberal rats. Dams were grouped into six; each group consists of six animals. Group 1-control treated with corn oil as vehicle, group II- 0.5mgPCBs/kg bw/day, group III- 0.5mgPCBs/kg bw/day with α-tocopherol (50mg α-tocopherol/kg bw/day), group IV- 1mgPCBs/kg bwt/day, group V- 1mgPCBs/kg bw/day with α-tocopherol (50mg α-tocopherol/kg bw/day) and group VI - α-tocopherol alone treated orally from postpartum day1-20. Male offspring rats were euthanized on post natal day 21. Testes were collected for the histological examination and Sertoli cell isolation. The protein levels of follicle-stimulating hormone receptor, androgen binding protein, androgen receptor, estrogen receptor α & β, Inhibin-β, transferrin, claudin-11, occludin, E-cadherin, connexin-43, c-fos, c-jun, SF1, USF1 & 2 were studied using western blot method. The testicular architecture was affected in the PCBs exposed rats but this effect was restored by α-tocopherol supplementation. PCBs decreased the protein levels of FSHR, AR, ABP, ERα & β, transferrin, claudin-11, occludin, E-cadherin, connexin-43, c-fos, c-jun, SF1, USF1 & 2 whereas inhibin-β protein level was found to be increased in Sertoli cells. These results suggested that α-tocopherol has ameliorative role against PCBs induced testicular Sertoli cell dysfunction in F1 progeny.
Collapse
Affiliation(s)
- Sugantha Priya Elayapillai
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600 113, Tamil Nadu, India
| | - Dhanaraj Teekaraman
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600 113, Tamil Nadu, India
| | - Raja Singh Paulraj
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600 113, Tamil Nadu, India
| | - Arunakaran Jagadeesan
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, 600 113, Tamil Nadu, India; Meenakshi Academy of Higher Education and Research, West KK Nagar, Chennai, 600 078, Tamil Nadu, India.
| |
Collapse
|
56
|
Nissinen L, Siljamäki E, Riihilä P, Piipponen M, Farshchian M, Kivisaari A, Kallajoki M, Raiko L, Peltonen J, Peltonen S, Kähäri VM. Expression of claudin-11 by tumor cells in cutaneous squamous cell carcinoma is dependent on the activity of p38δ. Exp Dermatol 2017; 26:771-777. [PMID: 27992079 DOI: 10.1111/exd.13278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
The incidence of cutaneous squamous cell carcinoma (cSCC) is rapidly increasing, and the prognosis of patients with metastatic disease is poor. There is an emerging need to identify molecular markers for predicting aggressive behaviour of cSCC. Here, we have examined the role of tight junction (TJ) components in the progression of cSCC. The expression pattern of mRNAs for TJ components was determined with RNA sequencing and oligonucleotide array-based expression analysis from cSCC cell lines (n=8) and normal human epidermal keratinocytes (NHEK, n=5). The expression of CLDN11 was specifically elevated in primary cSCC cell lines (n=5), but low or absent in metastatic cSCC cell lines (n=3) and NHEKs. Claudin-11 was detected in cell-cell contacts of primary cSCC cells in culture by indirect immunofluorescence analysis. Analysis of a large panel of tissue samples from sporadic UV-induced cSCC (n=65), cSCC in situ (n=56), actinic keratoses (n=31), seborrhoeic keratoses (n=7) and normal skin (n=16) by immunohistochemistry showed specific staining for claudin-11 in intercellular junctions of keratinizing tumor cells in well and moderately differentiated cSCCs, whereas no staining for claudin-11 was detected in poorly differentiated tumors. The expression of claudin-11 in cSCC cells was dependent on the activity of p38δ MAPK and knock-down of claudin-11 enhanced cSCC cell invasion. These findings provide evidence for the role of claudin-11 in regulation of cSCC invasion and suggest loss of claudin-11 expression in tumor cells as a biomarker for advanced stage of cSCC.
Collapse
Affiliation(s)
- Liisa Nissinen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Elina Siljamäki
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Pilvi Riihilä
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Minna Piipponen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Mehdi Farshchian
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Atte Kivisaari
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Laura Raiko
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha Peltonen
- Department of Cell Biology and Anatomy, University of Turku, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
| | - Veli-Matti Kähäri
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| |
Collapse
|
57
|
Peter A, Fatykhova D, Kershaw O, Gruber AD, Rueckert J, Neudecker J, Toennies M, Bauer TT, Schneider P, Schimek M, Eggeling S, Suttorp N, Hocke AC, Hippenstiel S. Localization and pneumococcal alteration of junction proteins in the human alveolar-capillary compartment. Histochem Cell Biol 2017; 147:707-719. [PMID: 28247028 DOI: 10.1007/s00418-017-1551-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2017] [Indexed: 02/03/2023]
Abstract
Loss of alveolar barrier function with subsequent respiratory failure is a hallmark of severe pneumonia. Although junctions between endo- and epithelial cells regulate paracellular fluid flux, little is known about their composition and regulation in the human alveolar compartment. High autofluorescence of human lung tissue in particular complicates the determination of subcellular protein localization. By comparing conventional channel mode confocal imaging with spectral imaging and linear unmixing, we demonstrate that background fluorescent spectra and fluorophore signals could be rigorously separated resulting in complete recovery of the specific signal at a high signal-to-noise ratio. Using this technique and Western blotting, we show the expression patterns of tight junction proteins occludin, ZO-1 as well as claudin-3, -4, -5 and -18 and adherence junction protein VE-cadherin in naive or Streptococcus pneumoniae-infected human lung tissue. In uninfected tissues, occludin and ZO-1 formed band-like structures in alveolar epithelial cells type I (AEC I), alveolar epithelial cells type II (AEC II) and lung capillaries, whereas claudin-3, -4 and -18 were visualised in AEC II. Claudin-5 was detected in the endothelium only. Claudin-3, -5, -18 displayed continuous band-like structures, while claudin-4 showed a dot-like expression. Pneumococcal infection reduced alveolar occludin, ZO-1, claudin-5 and VE-cadherin but did not change the presence of claudin-3, -4 and -18. Spectral confocal microscopy allows for the subcellular structural analysis of proteins in highly autofluorescent human lung tissue. The thereby observed deterioration of lung alveolar junctional organisation gives a structural explanation for alveolar barrier disruption in severe pneumococcal pneumonia.
Collapse
Affiliation(s)
- Andrea Peter
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Department for Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355, Berlin, Germany
| | - Diana Fatykhova
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Jens Rueckert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité-Universitätsmedizin Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Mario Toennies
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Torsten T Bauer
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany
| | - Paul Schneider
- Department for General and Thoracic Surgery, DRK Clinics, Drontheimer Strasse 39-40, 13359, Berlin, Germany
| | - Maria Schimek
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Rudower Straße 48, 12351, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas C Hocke
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
58
|
Male reproductive toxicity of CrVI: In-utero exposure to CrVI at the critical window of testis differentiation represses the expression of Sertoli cell tight junction proteins and hormone receptors in adult F 1 progeny rats. Reprod Toxicol 2017; 69:84-98. [PMID: 28192182 DOI: 10.1016/j.reprotox.2017.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/19/2017] [Accepted: 02/08/2017] [Indexed: 11/24/2022]
Abstract
The effect of gestational exposure to CrVI (occupational/environmental pollutant and target to Sertoli cells(SC)) was tested in a rat model during the testicular differentiation from the bipotential gonad may interrupt spermatogenesis by disrupting SC tight junctions(TJ) and it's proteins and hormone receptors. Pregnant Wistar rats were exposed to 50/100/200ppm CrVI through drinking water during embryonic days 9-14. On Postnatal day 120, testes were subjected to ion exchange chromatographic analysis and revealed increased level of CrIII in SCs and germ cells, serum and testicular interstitial fluid(TIF). Microscopic analyses showed seminiferous tubules atrophy and disruption of SC TJ, which also recorded decreased testosterone in TIF. mRNA and Protein expression analyses attested decreased level of Fshr, Ar, occludin and claudin-11 in SCs. Immunofluorescent detection revealed weak signal of TJ proteins. Taken together, we concluded that gestational exposure to CrVI interferes with the expression of SC TJ proteins due to attenuated expression of hormone receptors.
Collapse
|
59
|
Urban fine particulate matter exposure causes male reproductive injury through destroying blood-testis barrier (BTB) integrity. Toxicol Lett 2016; 266:1-12. [PMID: 27939690 DOI: 10.1016/j.toxlet.2016.12.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 11/21/2022]
Abstract
Blood-testis barrier (BTB) provides a suitable microenvironment for germ cells that is required for spermatogenesis. Exposure to particulate matter (PM) is recognized to occasion male reproductive impairment, but the mechanism of which remains unclear. Male Sprague-Dawley (SD) rats were used to establish animal models with PM2.5 exposure concentration of 0, 10, and 20mg/kg.b.w. once a day for four weeks. Success rate of mating, sperm quality, epididymal morphology, expressions of spermatogenesis markers, superoxide dismutases (SOD) activity and expression in testicular tissues, and expressions of BTB junction proteins were detected. In addition, in vitro experiments were also performed. After PM2.5 treatment, reactive oxygen species (ROS) production and apoptosis of Sertoli cells were analyzed. Our results indicated that after PM2.5 exposure male rats presented inferior uberty and sperm quality, with decreased expressions of spermatogenesis markers, escalated SOD activity and expression levels, and reduced expressions of tight junction, adherens junction, and gap junction proteins in testicular tissues. Meantime, PM2.5-treated Sertoli cells displayed increased SOD production and apoptosis. PM2.5 exposure engenders male reproductive function injury through breaking BTB integrity.
Collapse
|
60
|
Characterization of the Tetraspan Junctional Complex (4JC) superfamily. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1859:402-414. [PMID: 27916633 DOI: 10.1016/j.bbamem.2016.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/22/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022]
Abstract
Connexins or innexins form gap junctions, while claudins and occludins form tight junctions. In this study, statistical data, derived using novel software, indicate that these four junctional protein families and eleven other families of channel and channel auxiliary proteins are related by common descent and comprise the Tetraspan (4 TMS) Junctional Complex (4JC) Superfamily. These proteins all share similar 4 transmembrane α-helical (TMS) topologies. Evidence is presented that they arose via an intragenic duplication event, whereby a 2 TMS-encoding genetic element duplicated tandemly to give 4 TMS proteins. In cases where high resolution structural data were available, the conclusion of homology was supported by conducting structural comparisons. Phylogenetic trees reveal the probable relationships of these 15 families to each other. Long homologues containing fusions to other recognizable domains as well as internally duplicated or fused domains are reported. Large "fusion" proteins containing 4JC domains proved to fall predominantly into family-specific patterns as follows: (1) the 4JC domain was N-terminal; (2) the 4JC domain was C-terminal; (3) the 4JC domain was duplicated or occasionally triplicated and (4) mixed fusion types were present. Our observations provide insight into the evolutionary origins and subfunctions of these proteins as well as guides concerning their structural and functional relationships.
Collapse
|
61
|
Regulation of steroid hormones and energy status with cysteamine and its effect on spermatogenesis. Toxicol Appl Pharmacol 2016; 313:149-158. [DOI: 10.1016/j.taap.2016.10.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/23/2023]
|
62
|
Changes in Inflammatory Cytokines Accompany Deregulation of Claudin-11, Resulting in Inter-Sertoli Tight Junctions in Varicocele Rat Testes. J Urol 2016; 196:1303-12. [PMID: 27164517 DOI: 10.1016/j.juro.2016.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2016] [Indexed: 11/22/2022]
Abstract
PURPOSE To elucidate the changes that occur in the blood-testis barrier during varicocele we examined changes in Cldn11 (claudin-11), an element of the blood-testis barrier, as well as steroidogenesis and proinflammatory cytokines in a model of varicocele rat testes. MATERIALS AND METHODS Male rats with experimentally induced varicocele were sacrificed 4 weeks after operation. Testicular histology and blood testosterone concentrations were examined. The expression of tight junctions, steroidogenic enzymes, apoptosis and immune cell markers, and proinflammatory cytokines in the testes were evaluated by reverse transcriptase-polymerase chain reaction, Western blot and immunohistochemistry. RESULTS Weight and Johnsen scores of varicocele testes were lower than those of normal testes. mRNA expression of Bad and Bax increased whereas Bcl-xl and Bcl2 mRNA decreased in varicocele testes compared to controls. Although blood testosterone did not change, Leydig cell 3βHsd immunoreactivity, testicular 3βHsd6 and 17βHsd3 mRNA were significantly decreased in varicocele testes. Cldn11 mRNA and protein levels in varicocele testes were higher than in normal testes together with altered expression of Ocln, Zo1 and N-cadherin mRNA. Cldn11 immunoreactivity appeared as wavy strands at the periphery of the seminiferous epithelium in normal testes but was frequently found in the Sertoli cell cytoplasm in varicocele testes. In varicocele testes Tnfα, Il1α, Il6, Cd45, Cd3g and Cd3d mRNA was increased. CONCLUSIONS An increase in proinflammatory cytokines might be responsible for deregulation of Cldn11 in the Sertoli cells in varicocele testes, leading to alterations in the permeability of the blood-testis barrier and immunological barriers to normal spermatogenesis.
Collapse
|
63
|
Bulldan A, Dietze R, Shihan M, Scheiner-Bobis G. Non-classical testosterone signaling mediated through ZIP9 stimulates claudin expression and tight junction formation in Sertoli cells. Cell Signal 2016; 28:1075-85. [PMID: 27164415 DOI: 10.1016/j.cellsig.2016.04.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 11/27/2022]
Abstract
In the classical signaling pathway, testosterone regulates gene expression by activating the cytosolic/nuclear androgen receptor. In the non-classical pathway, testosterone activates cytosolic signaling cascades that are normally triggered by growth factors. The nature of the receptor involved in this signaling pathway is a source of controversy. In the Sertoli cell line 93RS2, which lacks the classical AR, we determined that testosterone stimulates the non-classical signaling pathway, characterized by the phosphorylation of Erk1/2 and transcription factors CREB and ATF-1. We also demonstrated that testosterone increases the expression of the tight junction (TJ) proteins claudin-1 and claudin-5. Both of these proteins are known to be essential constituents of TJs between Sertoli cells, and as a consequence of their increased expression transepithelial resistance across Sertoli cell monolayers is increased. ZIP9 is a Zn(2+)transporter that was recently shown to be a membrane-bound testosterone receptor. Silencing its expression in 93RS2 Sertoli cells by siRNA completely prevents Erk1/2, CREB, and ATF-1 phosphorylation as well the stimulation of claudin-1 and -5 expression and TJ formation between neighboring cells. The study presented here demonstrates for the first time that in Sertoli cells testosterone acts through the receptor ZIP9 to trigger the non-classical signaling cascade, resulting in increased claudin expression and TJ formation. Since TJ formation is a prerequisite for the maintenance of the blood-testis barrier, the testosterone/ZIP9 effects might be significant for male physiology. Further assessment of these interactions will help to supplement our knowledge concerning the mechanism by which testosterone plays a role in male fertility.
Collapse
Affiliation(s)
- Ahmed Bulldan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany
| | - Raimund Dietze
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany
| | - Mazen Shihan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin Justus-Liebig-Universität, Giessen, Germany.
| |
Collapse
|
64
|
Use of a rat ex-vivo testis culture method to assess toxicity of select known male reproductive toxicants. Reprod Toxicol 2016; 60:92-103. [DOI: 10.1016/j.reprotox.2016.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 01/21/2023]
|
65
|
Papadopoulos D, Dietze R, Shihan M, Kirch U, Scheiner-Bobis G. Dehydroepiandrosterone Sulfate Stimulates Expression of Blood-Testis-Barrier Proteins Claudin-3 and -5 and Tight Junction Formation via a Gnα11-Coupled Receptor in Sertoli Cells. PLoS One 2016; 11:e0150143. [PMID: 26938869 PMCID: PMC4777551 DOI: 10.1371/journal.pone.0150143] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/09/2016] [Indexed: 11/18/2022] Open
Abstract
Dehydroepiandrosterone sulfate (DHEAS) is a circulating sulfated steroid considered to be a pro-androgen in mammalian physiology. Here we show that at a physiological concentration (1 μM), DHEAS induces the phosphorylation of the kinase Erk1/2 and of the transcription factors CREB and ATF-1 in the murine Sertoli cell line TM4. This signaling cascade stimulates the expression of the tight junction (TJ) proteins claudin-3 and claudin-5. As a consequence of the increased expression, tight junction connections between neighboring Sertoli cells are augmented, as demonstrated by measurements of transepithelial resistance. Phosphorylation of Erk1/2, CREB, or ATF-1 is not affected by the presence of the steroid sulfatase inhibitor STX64. Erk1/2 phosphorylation was not observed when dehydroepiandrosterone (DHEA) was used instead of DHEAS. Abrogation of androgen receptor (AR) expression by siRNA did not affect DHEAS-stimulated Erk1/2 phosphorylation, nor did it change DHEAS-induced stimulation of claudin-3 and claudin-5 expression. All of the above indicate that desulfation and conversion of DHEAS into a different steroid hormone is not required to trigger the DHEAS-induced signaling cascade. All activating effects of DHEAS, however, are abolished when the expression of the G-protein Gnα11 is suppressed by siRNA, including claudin-3 and -5 expression and TJ formation between neighboring Sertoli cells as indicated by reduced transepithelial resistance. Taken together, these results are consistent with the effects of DHEAS being mediated through a membrane-bound G-protein-coupled receptor interacting with Gnα11 in a signaling pathway that resembles the non-classical signaling pathways of steroid hormones. Considering the fact that DHEAS is produced in reproductive organs, these findings also suggest that DHEAS, by acting as an autonomous steroid hormone and influencing the formation and dynamics of the TJ at the blood-testis barrier, might play a crucial role for the regulation and maintenance of male fertility.
Collapse
Affiliation(s)
- Dimitrios Papadopoulos
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Raimund Dietze
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Mazen Shihan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Ulrike Kirch
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
66
|
AKAP9, a Regulator of Microtubule Dynamics, Contributes to Blood-Testis Barrier Function. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:270-84. [PMID: 26687990 DOI: 10.1016/j.ajpath.2015.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/02/2015] [Accepted: 10/13/2015] [Indexed: 01/23/2023]
Abstract
The blood-testis barrier (BTB), formed between adjacent Sertoli cells, undergoes extensive remodeling to facilitate the transport of preleptotene spermatocytes across the barrier from the basal to apical compartments of the seminiferous tubules for further development and maturation into spermatozoa. The actin cytoskeleton serves unique structural and supporting roles in this process, but little is known about the role of microtubules and their regulators during BTB restructuring. The large isoform of the cAMP-responsive scaffold protein AKAP9 regulates microtubule dynamics and nucleation at the Golgi. We found that conditional deletion of Akap9 in mice after the initial formation of the BTB at puberty leads to infertility. Akap9 deletion results in marked alterations in the organization of microtubules in Sertoli cells and a loss of barrier integrity despite a relatively intact, albeit more apically localized F-actin and BTB tight junctional proteins. These changes are accompanied by a loss of haploid spermatids due to impeded meiosis. The barrier, however, progressively reseals in older Akap9 null mice, which correlates with a reduction in germ cell apoptosis and a greater incidence of meiosis. However, spermiogenesis remains defective, suggesting additional roles for AKAP9 in this process. Together, our data suggest that AKAP9 and, by inference, the regulation of the microtubule network are critical for BTB function and subsequent germ cell development during spermatogenesis.
Collapse
|
67
|
Lucke-Wold BP, Logsdon AF, Smith KE, Turner RC, Alkon DL, Tan Z, Naser ZJ, Knotts CM, Huber JD, Rosen CL. Bryostatin-1 Restores Blood Brain Barrier Integrity following Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2015; 52:1119-1134. [PMID: 25301233 PMCID: PMC5000781 DOI: 10.1007/s12035-014-8902-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023]
Abstract
Recent wars in Iraq and Afghanistan have accounted for an estimated 270,000 blast exposures among military personnel. Blast traumatic brain injury (TBI) is the 'signature injury' of modern warfare. Blood brain barrier (BBB) disruption following blast TBI can lead to long-term and diffuse neuroinflammation. In this study, we investigate for the first time the role of bryostatin-1, a specific protein kinase C (PKC) modulator, in ameliorating BBB breakdown. Thirty seven Sprague-Dawley rats were used for this study. We utilized a clinically relevant and validated blast model to expose animals to moderate blast exposure. Groups included: control, single blast exposure, and single blast exposure + bryostatin-1. Bryostatin-1 was administered i.p. 2.5 mg/kg after blast exposure. Evan's blue, immunohistochemistry, and western blot analysis were performed to assess injury. Evan's blue binds to albumin and is a marker for BBB disruption. The single blast exposure caused an increase in permeability compared to control (t = 4.808, p < 0.05), and a reduction back toward control levels when bryostatin-1 was administered (t = 5.113, p < 0.01). Three important PKC isozymes, PKCα, PKCδ, and PKCε, were co-localized primarily with endothelial cells but not astrocytes. Bryostatin-1 administration reduced toxic PKCα levels back toward control levels (t = 4.559, p < 0.01) and increased the neuroprotective isozyme PKCε (t = 6.102, p < 0.01). Bryostatin-1 caused a significant increase in the tight junction proteins VE-cadherin, ZO-1, and occludin through modulation of PKC activity. Bryostatin-1 ultimately decreased BBB breakdown potentially due to modulation of PKC isozymes. Future work will examine the role of bryostatin-1 in preventing chronic neurodegeneration following repetitive neurotrauma.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Aric F Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Kelly E Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, 26506, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Office of Professional Studies in Health Sciences, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Chelsea M Knotts
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jason D Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Charles L Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV, 26506-9183, USA.
| |
Collapse
|
68
|
Gerber J, Heinrich J, Brehm R. Blood-testis barrier and Sertoli cell function: lessons from SCCx43KO mice. Reproduction 2015; 151:R15-27. [PMID: 26556893 DOI: 10.1530/rep-15-0366] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/10/2015] [Indexed: 01/23/2023]
Abstract
The gap junction protein connexin43 (CX43) plays a vital role in mammalian spermatogenesis by allowing for direct cytoplasmic communication between neighbouring testicular cells. In addition, different publications suggest that CX43 in Sertoli cells (SC) might be important for blood-testis barrier (BTB) formation and BTB homeostasis. Thus, through the use of the Cre-LoxP recombination system, a transgenic mouse line was developed in which only SC are deficient of the gap junction protein, alpha 1 (Gja1) gene. Gja1 codes for the protein CX43. This transgenic mouse line has been commonly defined as the SC specific CX43 knockout (SCCx43KO) mouse line. Within the seminiferous tubule, SC aid in spermatogenesis by nurturing germ cells and help them to proliferate and mature. Owing to the absence of CX43 within the SC, homozygous KO mice are infertile, have reduced testis size, and mainly exhibit spermatogenesis arrest at the level of spermatogonia, seminiferous tubules containing only SC (SC-only syndrome) and intratubular SC-clusters. Although the SC specific KO of CX43 does not seem to have an adverse effect on BTB integrity, CX43 influences BTB composition as the expression pattern of different BTB proteins (like OCCLUDIN, β-CATENIN, N-CADHERIN, and CLAUDIN11) is altered in mutant males. The supposed roles of CX43 in dynamic BTB regulation, BTB assembly and/or disassembly and its possible interaction with other junctional proteins composing this unique barrier are discussed. Data collectively indicate that CX43 might represent an important regulator of dynamic BTB formation, composition and function.
Collapse
Affiliation(s)
- Jonathan Gerber
- Institute of AnatomyUniversity of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hannover, Germany
| | - Julia Heinrich
- Institute of AnatomyUniversity of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hannover, Germany
| | - Ralph Brehm
- Institute of AnatomyUniversity of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hannover, Germany
| |
Collapse
|
69
|
Mruk DD, Cheng CY. The Mammalian Blood-Testis Barrier: Its Biology and Regulation. Endocr Rev 2015; 36:564-91. [PMID: 26357922 PMCID: PMC4591527 DOI: 10.1210/er.2014-1101] [Citation(s) in RCA: 442] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/03/2015] [Indexed: 12/31/2022]
Abstract
Spermatogenesis is the cellular process by which spermatogonia develop into mature spermatids within seminiferous tubules, the functional unit of the mammalian testis, under the structural and nutritional support of Sertoli cells and the precise regulation of endocrine factors. As germ cells develop, they traverse the seminiferous epithelium, a process that involves restructuring of Sertoli-germ cell junctions, as well as Sertoli-Sertoli cell junctions at the blood-testis barrier. The blood-testis barrier, one of the tightest tissue barriers in the mammalian body, divides the seminiferous epithelium into 2 compartments, basal and adluminal. The blood-testis barrier is different from most other tissue barriers in that it is not only comprised of tight junctions. Instead, tight junctions coexist and cofunction with ectoplasmic specializations, desmosomes, and gap junctions to create a unique microenvironment for the completion of meiosis and the subsequent development of spermatids into spermatozoa via spermiogenesis. Studies from the past decade or so have identified the key structural, scaffolding, and signaling proteins of the blood-testis barrier. More recent studies have defined the regulatory mechanisms that underlie blood-testis barrier function. We review here the biology and regulation of the mammalian blood-testis barrier and highlight research areas that should be expanded in future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- Center for Biomedical Research, Population Council, New York, New York 10065
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, New York, New York 10065
| |
Collapse
|
70
|
Jiang XH, Bukhari I, Zheng W, Yin S, Wang Z, Cooke HJ, Shi QH. Blood-testis barrier and spermatogenesis: lessons from genetically-modified mice. Asian J Androl 2015; 16:572-80. [PMID: 24713828 PMCID: PMC4104086 DOI: 10.4103/1008-682x.125401] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The blood-testis barrier (BTB) is found between adjacent Sertoli cells in the testis where it creates a unique microenvironment for the development and maturation of meiotic and postmeiotic germ cells in seminiferous tubes. It is a compound proteinous structure, composed of several types of cell junctions including tight junctions (TJs), adhesion junctions and gap junctions (GJs). Some of the junctional proteins function as structural proteins of BTB and some have regulatory roles. The deletion or functional silencing of genes encoding these proteins may disrupt the BTB, which may cause immunological or other damages to meiotic and postmeiotic cells and ultimately lead to spermatogenic arrest and infertility. In this review, we will summarize the findings on the BTB structure and function from genetically-modified mouse models and discuss the future perspectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qing-Hua Shi
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China; Institute of Physics, Chinese Academy of Sciences, Hefei, China,
| |
Collapse
|
71
|
Wang XP, Lei F, Du F, Chai YS, Jiang JF, Wang YG, Yu X, Yan XJ, Xing DM, Du LJ. Protection of Gastrointestinal Mucosa from Acute Heavy Alcohol Consumption: The Effect of Berberine and Its Correlation with TLR2, 4/IL1β-TNFα Signaling. PLoS One 2015. [PMID: 26226164 PMCID: PMC4520689 DOI: 10.1371/journal.pone.0134044] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The purpose of the present study is to confirm the protective effect of berberine (BBR) on gastrointestinal injury caused by acute heavy alcohol exposure, an effect that has not been reported previously. Our research details how BBR protects against gastrointestinal injuries from acute alcohol exposure using both in vivo and in vitro experiments. Acute high alcohol concentrations lead to obvious damage to the gastrointestinal mucosa, resulting in necrosis of the intestinal mucosa. Oral administration of BBR was able to significantly reduce this alcohol-induced damage, inhibit increases of alcohol-induced TNFα and IL-1β expression in gastrointestinal mucosa as well as their upstream signals TLR2 and TLR4, and regulate cytokines that modulate tight junctions. Alcohol consumption is a popular human social behavior worldwide, and the present study reports a comprehensive mechanism by which BBR protects against gastrointestinal injuries from alcohol stress, providing people with a novel application of BBR.
Collapse
Affiliation(s)
- Xin-Pei Wang
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fan Lei
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Feng Du
- Department of Mathematics, Tulane University, New Orleans, LA, 70118, United States of America
| | - Yu-Shuang Chai
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jing-Fei Jiang
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yu-Gang Wang
- MD Anderson Cancer Center, University of Texas, Houston, Texas, 77030, United States of America
| | - Xuan Yu
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiao-Jin Yan
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dong-Ming Xing
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Li-Jun Du
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- * E-mail:
| |
Collapse
|
72
|
Ahmed E, Nagaoka K, Fayez M, Abdel-Daim MM, Samir H, Watanabe G. Suppressive effects of long-term exposure to P-nitrophenol on gonadal development, hormonal profile with disruption of tissue integrity, and activation of caspase-3 in male Japanese quail (Coturnix japonica). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:10930-42. [PMID: 25772865 PMCID: PMC4490174 DOI: 10.1007/s11356-015-4245-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/17/2015] [Indexed: 06/03/2023]
Abstract
P-Nitrophenol (PNP) is considered to be one of nitrophenol derivatives of diesel exhaust particles. PNP is a major metabolite of some organophosphorus compounds. PNP is a persistent organic pollutant as well as one of endocrine-disrupting compounds. Consequently, bioaccumulation of PNP potentiates toxicity. The objectives of the current study were to assess in vivo adverse effects of long-term low doses of PNP exposure on reproductive system during development stage. Twenty-eight-day-old male Japanese quails were orally administered different doses of PNP (0, 0.01, 0.1, 1 mg/kg body weight) daily for 2.5 months. Testicular histopathology, hormones, caspase-3 (CASP3), and claudin-1 (CLDN1) tight junction protein, as well as plasma hormones were analyzed. The results revealed that long-term PNP exposure caused testicular histopathological changes such as vacuolation of spermatogenic cell and spermatocyte with significant testicular and cloacal gland atrophy. PNP activated CASP3 enzyme that is an apoptosis-related cysteine peptidase. Besides, it disrupted the expression of CLDN1. Furthermore, a substantial decrease in plasma concentrations of luteinizing hormone (LH) and testosterone was observed after 2 and 2.5 months in the PNP-treated groups. Meanwhile, the pituitary LH did not significantly change. Site of action of PNP may be peripheral on testicular development and/or centrally on the hypothalamic-pituitary-gonadal axis through reduction of pulsatile secretion of gonadotrophin-releasing hormone. Consequently, it may reduce the sensitivity of the anterior pituitary gland to secrete LH. In conclusion, PNP induced profound endocrine disruption in the form of hormonal imbalance, induction of CASP3, and disruption of CLDN1 expression in the testis. Hence, it may hinder the reproductive processes.
Collapse
Affiliation(s)
- Eman Ahmed
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan,
| | | | | | | | | | | |
Collapse
|
73
|
Abstract
PURPOSE OF REVIEW The present review examines the role of actin binding proteins (ABPs) on blood-testis barrier (BTB), an androgen-dependent ultrastructure in the testis, in particular their involvement on BTB remodeling during spermatogenesis. RECENT FINDINGS The BTB divides the seminiferous epithelium into the basal and the adluminal compartments. The BTB is constituted by coexisting actin-based tight junction, basal ectoplasmic specialization, and gap junction, and also intermediate filament-based desmosome between Sertoli cells near the basement membrane. Junctions at the BTB undergo continuous remodeling to facilitate the transport of preleptotene spermatocytes residing in the basal compartment across the immunological barrier during spermatogenesis. Thus, meiosis I/II and postmeiotic spermatid development take place in the adluminal compartment behind the BTB. BTB remodeling also regulates exchanges of biomolecules between the two compartments. As tight junction, basal ectoplasmic specialization, and gap junction use F-actin for attachment, actin microfilaments rapidly convert between their bundled and unbundled/branched configuration to confer BTB plasticity. The events of actin reorganization are regulated by two major classes of ABPs that convert actin microfilaments between their bundled and branched/unbundled configuration. SUMMARY We provide a model on how ABPs regulate BTB remodeling, shedding new light on unexplained male infertility, such as environmental toxicant-induced reproductive dysfunction since the testis, in particular the BTB, is sensitive to environmental toxicants, such as cadmium, bisphenol A, phthalates, and PFOS (perfluorooctanesulfonic acid or perfluorooctane sulfonate).
Collapse
Affiliation(s)
- Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | | | | |
Collapse
|
74
|
Welborn JP, Davis MG, Ebers SD, Stodden GR, Hayashi K, Cheatwood JL, Rao MK, MacLean JA. Rhox8 Ablation in the Sertoli Cells Using a Tissue-Specific RNAi Approach Results in Impaired Male Fertility in Mice. Biol Reprod 2015; 93:8. [PMID: 25972016 DOI: 10.1095/biolreprod.114.124834] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 05/11/2015] [Indexed: 12/27/2022] Open
Abstract
The reproductive homeobox X-linked, Rhox, genes encode transcription factors that are selectively expressed in reproductive tissues. While there are 33 Rhox genes in mice, only Rhox and Rhox8 are expressed in Sertoli cells, suggesting that they may regulate the expression of somatic-cell gene products crucial for germ cell development. We previously characterized Rhox5-null mice, which are subfertile, exhibiting excessive germ cell apoptosis and compromised sperm motility. To assess the role of Rhox8 in Sertoli cells, we used a tissue-specific RNAi approach to knockdown RHOX8 in vivo, in which the Rhox5 promoter was used to drive Rhox8-siRNA transgene expression in the postnatal Sertoli cells. Western and immunohistochemical analysis confirmed Sertoli-specific knockdown of RHOX8. However, other Sertoli markers, Gata1 and Rhox5, maintained normal expression patterns, suggesting that the knockdown was specific. Interestingly, male RHOX8-knockdown animals showed significantly reduced spermatogenic output, increased germ cell apoptosis, and compromised sperm motility, leading to impaired fertility. Importantly, our results revealed that while some RHOX5-dependent factors were also misregulated in Sertoli cells of RHOX8-knockdown animals, the majority were not, and novel putative RHOX8-regulated genes were identified. This suggests that while reduction in levels of RHOX5 and RHOX8 in Sertoli cells elicits similar phenotypes, these genes are not entirely redundant. Taken together, our study underscores the importance of Rhox genes in male fertility and suggests that Sertoli cell-specific expression of Rhox5 and Rhox8 is critical for complete male fertility.
Collapse
Affiliation(s)
- Joshua P Welborn
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Matthew G Davis
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Steven D Ebers
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Genna R Stodden
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Joseph L Cheatwood
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Manjeet K Rao
- Department of Cellular and Structural Biology, Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - James A MacLean
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| |
Collapse
|
75
|
Dietze R, Shihan M, Stammler A, Konrad L, Scheiner-Bobis G. Cardiotonic steroid ouabain stimulates expression of blood-testis barrier proteins claudin-1 and -11 and formation of tight junctions in Sertoli cells. Mol Cell Endocrinol 2015; 405:1-13. [PMID: 25666991 DOI: 10.1016/j.mce.2015.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 01/03/2023]
Abstract
The interaction of ouabain with the sodium pump induces signalling cascades resembling those triggered by hormone/receptor interactions. In the rat Sertoli cell line 93RS2, ouabain at low concentrations stimulates the c-Src/c-Raf/Erk1/2 signalling cascade via its interaction with the α4 isoform of the sodium pump expressed in these cells, leading to the activation of the transcription factor CREB. As a result of this signalling sequence, ouabain stimulates expression of claudin-1 and claudin-11, which are also controlled by a CRE promoter. Both of these proteins are known to be essential constituents of tight junctions (TJ) between Sertoli cells, and as a result of the ouabain-induced signalling TJ formation between neighbouring Sertoli cells is significantly enhanced by the steroid. Thus, ouabain-treated cell monolayers display higher transepithelial resistance and reduced free diffusion of FITC-coupled dextran in tracer diffusion assays. Taking into consideration that the formation of TJ is indispensable for the maintenance of the blood-testis barrier (BTB) and therefore for male fertility, the actions of ouabain described here and the fact that this and other related cardiotonic steroids (CTS) are produced endogenously suggest a direct influence of ouabain/sodium pump interactions on the maintenance of the BTB and thereby an effect on male fertility. Since claudin-1 and claudin-11 are also present in other blood-tissue barriers, one can speculate that ouabain and perhaps other CTS influence the dynamics of these barriers as well.
Collapse
Affiliation(s)
- Raimund Dietze
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität, Giessen, Germany
| | - Mazen Shihan
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität, Giessen, Germany
| | - Angelika Stammler
- Zentrum f. Frauenheilkunde und Geburtshilfe, Justus-Liebig-Universität, Giessen, Germany; Institut f. Anatomie und Zellbiologie, Fachbereich Medizin, Justus-Liebig-Universität, Giessen, Germany
| | - Lutz Konrad
- Zentrum f. Frauenheilkunde und Geburtshilfe, Justus-Liebig-Universität, Giessen, Germany
| | - Georgios Scheiner-Bobis
- Institut für Veterinär-Physiologie und -Biochemie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität, Giessen, Germany.
| |
Collapse
|
76
|
Brøchner CB, Holst CB, Møllgård K. Outer brain barriers in rat and human development. Front Neurosci 2015; 9:75. [PMID: 25852456 PMCID: PMC4360706 DOI: 10.3389/fnins.2015.00075] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/20/2015] [Indexed: 12/24/2022] Open
Abstract
Complex barriers at the brain's surface, particularly in development, are poorly defined. In the adult, arachnoid blood-cerebrospinal fluid (CSF) barrier separates the fenestrated dural vessels from the CSF by means of a cell layer joined by tight junctions. Outer CSF-brain barrier provides diffusion restriction between brain and subarachnoid CSF through an initial radial glial end feet layer covered with a pial surface layer. To further characterize these interfaces we examined embryonic rat brains from E10 to P0 and forebrains from human embryos and fetuses (6–21st weeks post-conception) and adults using immunohistochemistry and confocal microscopy. Antibodies against claudin-11, BLBP, collagen 1, SSEA-4, MAP2, YKL-40, and its receptor IL-13Rα2 and EAAT1 were used to describe morphological characteristics and functional aspects of the outer brain barriers. Claudin-11 was a reliable marker of the arachnoid blood-CSF barrier. Collagen 1 delineated the subarachnoid space and stained pial surface layer. BLBP defined radial glial end feet layer and SSEA-4 and YKL-40 were present in both leptomeningeal cells and end feet layer, which transformed into glial limitans. IL-13Rα2 and EAAT1 were present in the end feet layer illustrating transporter/receptor presence in the outer CSF-brain barrier. MAP2 immunostaining in adult brain outlined the lower border of glia limitans; remnants of end feet were YKL-40 positive in some areas. We propose that outer brain barriers are composed of at least 3 interfaces: blood-CSF barrier across arachnoid barrier cell layer, blood-CSF barrier across pial microvessels, and outer CSF-brain barrier comprising glial end feet layer/pial surface layer.
Collapse
Affiliation(s)
- Christian B Brøchner
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| | - Camilla B Holst
- Department of Oncology, Copenhagen University Hospital Copenhagen, Denmark
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| |
Collapse
|
77
|
Park CJ, Ha CM, Lee JE, Gye MC. Claudin 11 inter-sertoli tight junctions in the testis of the korean soft-shelled turtle (Pelodiscus maackii). Biol Reprod 2015; 92:96. [PMID: 25761591 DOI: 10.1095/biolreprod.114.117804] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/03/2015] [Indexed: 11/01/2022] Open
Abstract
Expression of claudin 11 (CLDN11), a tight junction (TJ) protein, was examined in the Korean soft-shelled turtle (Pelodiscus maackii) testis. Spermatogenesis began during the breeding season and peaked at the end of the breeding season. Spermiation started in summer and peaked in autumn. The deduced amino acid sequence of P. maackii CLDN11 was similar to those of avian and mammalian species. During the nonbreeding season when spermatogenesis and testosterone production were active, testicular Cldn11 levels were high. In the seminiferous epithelium, strong, wavy CLDN11 strands parallel to the basement membrane delaminate the spermatogonia, and early spermatocytes are in the open compartment. Otherwise, CLDN11 was found beneath the early spermatocytes and in the Sertoli cell cytoplasm. Punctate zonula occludens 1 (ZO-1) immunoreactivity was found within the CLDN11 strands parallel to the basement membrane or at the outermost periphery of the seminiferous epithelium close to the basal lamina. During the breeding season, when circulating testosterone levels and spermatogenic activity was low, testicular CLDN11 level was lower than those during the nonbreeding season. CLDN11 was found at apicolateral contact sites between adjacent Sertoli cells devoid of the postmeiotic germ cells. At this time, lanthanum tracer diffused to the adluminal compartment of seminiferous epithelium. In cultured testis tissues, testosterone propionate significantly increased the level of Cldn11 mRNA. In P. maackii testis, CLDN11 participates in the development of the blood-testis barrier (BTB), where the CLDN11 expression was coupled with spermatogenic activity and circulating androgen levels, indicating the conserved nature of TJs expressing CLDN11 at the BTB in amniotes.
Collapse
Affiliation(s)
- Chan Jin Park
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Cheol Min Ha
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Jae Eun Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Myung Chan Gye
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| |
Collapse
|
78
|
D'Aurora M, Ferlin A, Di Nicola M, Garolla A, De Toni L, Franchi S, Palka G, Foresta C, Stuppia L, Gatta V. Deregulation of sertoli and leydig cells function in patients with Klinefelter syndrome as evidenced by testis transcriptome analysis. BMC Genomics 2015; 16:156. [PMID: 25879484 PMCID: PMC4362638 DOI: 10.1186/s12864-015-1356-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 02/19/2015] [Indexed: 01/01/2023] Open
Abstract
Background Klinefelter Syndrome (KS) is the most common abnormality of sex chromosomes (47,XXY) and represents the first genetic cause of male infertility. Mechanisms leading to KS testis degeneration are still not completely defined but considered to be mainly the result of germ cells loss. In order to unravel the molecular basis of global testis dysfunction in KS patients, we performed a transcriptome analysis on testis biopsies obtained from 6 azoospermic non-mosaic KS patients and 3 control subjects. Results The analysis found that, compared to controls, KS patients showed the differential up- and down-expression of 656 and 247 transcripts. The large majority of the deregulated transcripts were expressed by Sertoli cells (SCs) and Leydig cells (LCs). Functional analysis of the deregulated transcripts indicated changes of genes involved in cell death, inflammatory response, lipid metabolism, steroidogenesis, blood-testis-barrier formation and maintenance, as well as spermatogenesis failure. Conclusions Taken together, present data highlight the modulation of hundreds of genes in the somatic components of KS patient testis. The increased LCs steroidogenic function together with the impairment of inflammatory pathways and BTB structure, result in increased apoptosis. These findings may represent a critical roadmap for therapeutic intervention and prevention of KS-related testis failure. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1356-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marco D'Aurora
- Department of Psychological, Humanities and Territorial Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Via Dei Vestini 31, 66100, Chieti-Pescara, Italy. .,Functional Genetics Unit, Center of Excellence on Aging (Ce.S.I.), Via Dei Vestini 31, 66100, Chieti, Italy.
| | - Alberto Ferlin
- Department of Medicine, Section of Endocrinology and Centre for Human Reproduction Pathology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.
| | - Marta Di Nicola
- Department of Sperimental and Clinical Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Via dei Vestini 31, 66100, Chieti-Pescara, Italy.
| | - Andrea Garolla
- Department of Medicine, Section of Endocrinology and Centre for Human Reproduction Pathology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.
| | - Luca De Toni
- Department of Medicine, Section of Endocrinology and Centre for Human Reproduction Pathology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.
| | - Sara Franchi
- Department of Psychological, Humanities and Territorial Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Via Dei Vestini 31, 66100, Chieti-Pescara, Italy. .,Functional Genetics Unit, Center of Excellence on Aging (Ce.S.I.), Via Dei Vestini 31, 66100, Chieti, Italy.
| | - Giandomenico Palka
- Department of Oral Health and Biotechnological Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Via dei Vestini 31, 66100, Chieti-Pescara, Italy.
| | - Carlo Foresta
- Department of Medicine, Section of Endocrinology and Centre for Human Reproduction Pathology, University of Padova, Via Giustiniani 2, 35128, Padova, Italy.
| | - Liborio Stuppia
- Department of Psychological, Humanities and Territorial Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Via Dei Vestini 31, 66100, Chieti-Pescara, Italy. .,Functional Genetics Unit, Center of Excellence on Aging (Ce.S.I.), Via Dei Vestini 31, 66100, Chieti, Italy.
| | - Valentina Gatta
- Department of Psychological, Humanities and Territorial Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Via Dei Vestini 31, 66100, Chieti-Pescara, Italy. .,Functional Genetics Unit, Center of Excellence on Aging (Ce.S.I.), Via Dei Vestini 31, 66100, Chieti, Italy.
| |
Collapse
|
79
|
McMillan M, Andronicos N, Davey R, Stockwell S, Hinch G, Schmoelzl S. Claudin-8 expression in Sertoli cells and putative spermatogonial stem cells in the bovine testis. Reprod Fertil Dev 2015; 26:633-44. [PMID: 23673210 DOI: 10.1071/rd12259] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 04/16/2013] [Indexed: 12/25/2022] Open
Abstract
Adhesion molecules are expressed by both adult and embryonic stem cells, with different classes of adhesion molecules involved in cell-membrane and intercellular contacts. In this study the expression of the adhesion molecule claudin-8 (CLDN8), a tight-junction protein, was investigated as a potential marker for undifferentiated spermatogonia in the bovine testis. We found that CLDN8 was expressed by both spermatogonia and a subset of Sertoli cells in the bovine testis. We also showed co-expression of GFRα1 in testis cells with CLDN8 and with Dolichos biflorus agglutinin-fluorescein isothiocyanate (DBA-FITC) staining. We observed co-enrichment of spermatogonia and CLDN8-expressing Sertoli cells in DBA-FITC-assisted magnetic-activated cell sorting (MACS), an observation supported by results from fluorescence-activated cell sorting analysis, which showed CLDN8-expressing cells were over-represented in the MACS-positive cell fraction, leading to the hypothesis that CLDN8 may play a role in the spermatogonial stem-cell niche.
Collapse
Affiliation(s)
- Mary McMillan
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| | - Nicholas Andronicos
- CSIRO Animal, Food and Health Sciences, F. D. McMaster Laboratory, Armidale, NSW 2350, Australia
| | - Rhonda Davey
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| | - Sally Stockwell
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| | - Geoff Hinch
- School of Environmental and Rural Science, University of New England, Armidale, NSW 2351, Australia
| | - Sabine Schmoelzl
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| |
Collapse
|
80
|
Pérez CV, Theas MS, Jacobo PV, Jarazo-Dietrich S, Guazzone VA, Lustig L. Dual role of immune cells in the testis: Protective or pathogenic for germ cells? SPERMATOGENESIS 2014; 3:e23870. [PMID: 23687616 PMCID: PMC3644047 DOI: 10.4161/spmg.23870] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The purpose of this review is to describe how the immune cells present in the testis interact with the germinal epithelium contributing to survival or apoptosis of germ cells (GCs). Physiologically, the immunosuppressor testicular microenvironment protects GCs from immune attack, whereas in inflammatory conditions, tolerance is disrupted and immune cells and their mediators respond to GC self antigens, inducing damage of the germinal epithelium. Considering that experimental models of autoimmune orchitis have clarified the local immune mechanisms by which protection of the testis is compromised, we described the following topics in the testis of normal and orchitic rats: (1) cell adhesion molecule expression of seminiferous tubule specialized junctions and modulation of blood-testis barrier permeability by cytokines (2) phenotypic and functional characteristics of testicular dendritic cells, macrophages, effector and regulatory T cells and mast cells and (3) effects of pro-inflammatory cytokines (TNF-α, IL-6 and FasL) and the nitric oxide-nitric oxide synthase system on GC apoptosis.
Collapse
Affiliation(s)
- Cecilia V Pérez
- Instituto de Investigaciones Biomédicas; UBA/CONICET; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
81
|
Dando R, Pereira E, Kurian M, Barro-Soria R, Chaudhari N, Roper SD. A permeability barrier surrounds taste buds in lingual epithelia. Am J Physiol Cell Physiol 2014; 308:C21-32. [PMID: 25209263 DOI: 10.1152/ajpcell.00157.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epithelial tissues are characterized by specialized cell-cell junctions, typically localized to the apical regions of cells. These junctions are formed by interacting membrane proteins and by cytoskeletal and extracellular matrix components. Within the lingual epithelium, tight junctions join the apical tips of the gustatory sensory cells in taste buds. These junctions constitute a selective barrier that limits penetration of chemosensory stimuli into taste buds (Michlig et al. J Comp Neurol 502: 1003-1011, 2007). We tested the ability of chemical compounds to permeate into sensory end organs in the lingual epithelium. Our findings reveal a robust barrier that surrounds the entire body of taste buds, not limited to the apical tight junctions. This barrier prevents penetration of many, but not all, compounds, whether they are applied topically, injected into the parenchyma of the tongue, or circulating in the blood supply, into taste buds. Enzymatic treatments indicate that this barrier likely includes glycosaminoglycans, as it was disrupted by chondroitinase but, less effectively, by proteases. The barrier surrounding taste buds could also be disrupted by brief treatment of lingual tissue samples with DMSO. Brief exposure of lingual slices to DMSO did not affect the ability of taste buds within the slice to respond to chemical stimulation. The existence of a highly impermeable barrier surrounding taste buds and methods to break through this barrier may be relevant to basic research and to clinical treatments of taste.
Collapse
Affiliation(s)
- Robin Dando
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; and
| | - Elizabeth Pereira
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; and
| | - Mani Kurian
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; and
| | - Rene Barro-Soria
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; and
| | - Nirupa Chaudhari
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; and Program in Neuroscience, University of Miami, Miami, Florida
| | - Stephen D Roper
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; and Program in Neuroscience, University of Miami, Miami, Florida
| |
Collapse
|
82
|
Xiao X, Mruk DD, Tang EI, Massarwa R, Mok KW, Li N, Wong CKC, Lee WM, Snapper SB, Shilo BZ, Schejter ED, Cheng CY. N-wasp is required for structural integrity of the blood-testis barrier. PLoS Genet 2014; 10:e1004447. [PMID: 24967734 PMCID: PMC4072540 DOI: 10.1371/journal.pgen.1004447] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 05/02/2014] [Indexed: 01/06/2023] Open
Abstract
During spermatogenesis, the blood-testis barrier (BTB) segregates the adluminal (apical) and basal compartments in the seminiferous epithelium, thereby creating a privileged adluminal environment that allows post-meiotic spermatid development to proceed without interference of the host immune system. A key feature of the BTB is its continuous remodeling within the Sertoli cells, the major somatic component of the seminiferous epithelium. This remodeling is necessary to allow the transport of germ cells towards the seminiferous tubule interior, while maintaining intact barrier properties. Here we demonstrate that the actin nucleation promoting factor Neuronal Wiskott-Aldrich Syndrome Protein (N-WASP) provides an essential function necessary for BTB restructuring, and for maintaining spermatogenesis. Our data suggests that the N-WASP-Arp2/3 actin polymerization machinery generates branched-actin arrays at an advanced stage of BTB remodeling. These arrays are proposed to mediate the restructuring process through endocytic recycling of BTB components. Disruption of N-WASP in Sertoli cells results in major structural abnormalities to the BTB, including mis-localization of critical junctional and cytoskeletal elements, and leads to disruption of barrier function. These impairments result in a complete arrest of spermatogenesis, underscoring the critical involvement of the somatic compartment of the seminiferous tubules in germ cell maturation. Mammalian spermatogenesis takes place within a sheltered environment, whereby somatic Sertoli cells protect and guide germ cells as they mature and differentiate. A key structure generated by the protective Sertoli cell epithelium is the blood-testis barrier (BTB), a composite of junctional and cytoskeletal elements, which prevents exposure of post-meiotic spermatids to the immune system. The BTB is a highly dynamic structure, which needs to be dismantled and rapidly rebuilt, in order to allow passage of maturing preleptotene spermatocytes, without compromising their isolation. Here we show that N-WASP, a conserved facilitator of formation of branched actin microfilament arrays, provides a function that is essential for maintenance of an intact BTB. Genetic disruption of N-WASP in mouse Sertoli cells leads to loss of BTB impermeability, resulting in a complete arrest of spermatogenesis at early and post-meiotic stages. Based on the localization patterns of key elements, we propose that branched-actin filaments participate in recycling of BTB materials to ensure the dynamic and efficient maintenance of this structure, one of a series of blood-tissue barriers that preserve privileged organ environments.
Collapse
Affiliation(s)
- Xiang Xiao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Dolores D. Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Elizabeth I. Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - R'ada Massarwa
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ka Wai Mok
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Chris K. C. Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Will M. Lee
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Scott B. Snapper
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ben-Zion Shilo
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D. Schejter
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail: (EDS); (CYC)
| | - C. Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
- * E-mail: (EDS); (CYC)
| |
Collapse
|
83
|
Yang Q, Hao J, Chen M, Li G. Dermatopontin is a novel regulator of the CdCl2-induced decrease in claudin-11 expression. Toxicol In Vitro 2014; 28:1158-64. [PMID: 24909373 DOI: 10.1016/j.tiv.2014.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 05/08/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
Cadmium (Cd) is a ubiquitous environmental heavy metal, which may be harmful to the reproductive functions through injury to the blood-testis barrier (BTB). However, the underlying mechanism of this adverse effect on the BTB remains uncharacterized. A preliminary study revealed that dermatopontin (DPT) expression was significantly increased in Cd chloride (CdCl2)-treated Sertoli cells in vitro, which suggested that an increase in DPT expression is crucial for CdCl2-induced BTB damage. To explore this further, in the present study we initially determined that DPT is expressed in testis Sertoli cells. The treatment of cells with CdCl2 resulted in a significant increase in DPT expression and a parallel decrease in claudin-11 expression, both in vivo and in vitro. To confirm the relationship between DPT and claudin-11, a DPT-silenced 15P-1 Sertoli cell model was established. We determined that DPT silencing could partly reduce the CdCl2-induced decrease in claudin-11 expression. Additionally, western blot analyses demonstrated that the p38 signaling pathway is involved in the effect of CdCl2 on DPT expression. In conclusion, the present study provides the first evidence that DPT may be a novel effector of CdCl2, highlighting the significant role of DPT in the regulation of claudin-11 expression.
Collapse
Affiliation(s)
- Qiaozhen Yang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jie Hao
- The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Maoxin Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
84
|
Gilio JM, Portaro FC, Borella MI, Lameu C, Camargo AC, Alberto-Silva C. A bradykinin-potentiating peptide (BPP-10c) from Bothrops jararaca induces changes in seminiferous tubules. J Venom Anim Toxins Incl Trop Dis 2013; 19:28. [PMID: 24195771 PMCID: PMC4176135 DOI: 10.1186/1678-9199-19-28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 10/25/2013] [Indexed: 11/20/2022] Open
Abstract
Background The testis-specific isoform of angiotensin-converting enzyme (tACE) is exclusively expressed in germ cells during spermatogenesis. Although the exact role of tACE in male fertility is unknown, it clearly plays a critical function in spermatogenesis. The dipeptidase domain of tACE is identical to the C-terminal catalytic domain of somatic ACE (sACE). Bradykinin potentiating peptides (BPPs) from snake venoms are the first natural sACE inhibitors described and their structure–activity relationship studies were the basis for the development of antihypertensive drugs such as captopril. In recent years, it has been showed that a number of BPPs – including BPP-10c – are able to distinguish between the N- and C-active sites of sACE, what is not applicable to captopril. Considering the similarity between tACE and sACE (and since BPPs are able to distinguish between the two active sites of sACE), the effects of the BPP-10c and captopril on the structure and function of the seminiferous epithelium were characterized in the present study. BPP-10c and captopril were administered in male Swiss mice by intraperitoneal injection (4.7 μmol/kg for 15 days) and histological sections of testes were analyzed. Classification of seminiferous tubules and stage analysis were carried out for quantitative evaluation of germ cells of the seminiferous epithelium. The blood-testis barrier (BTB) permeability and distribution of claudin-1 in the seminiferous epithelium were analyzed by hypertonic fixative method and immunohistochemical analyses of testes, respectively. Results The morphology of seminiferous tubules from animals treated with BPP-10c showed an intense disruption of the epithelium, presence of atypical multinucleated cells in the lumen and degenerated germ cells in the adluminal compartment. BPP-10c led to an increase in the number of round spermatids and total support capacity of Sertoli cell in stages I, V, VII/VIII of the seminiferous epithelium cycle, without affecting BTB permeability and the distribution of claudin-1 in the seminiferous epithelium. Interestingly, no morphological or morphometric alterations were observed in animals treated with captopril. Conclusions The major finding of the present study was that BPP-10c, and not captopril, modifies spermatogenesis by causing hyperplasia of round spermatids in stages I, V, and VII/VIII of the spermatogenic cycle.
Collapse
Affiliation(s)
| | | | | | | | | | - Carlos Alberto-Silva
- Natural and Human Sciences Center (CCNH), Federal University of ABC (UFABC), Santo André, São Paulo State, Brazil.
| |
Collapse
|
85
|
Mok KW, Mruk DD, Cheng CY. Regulation of blood-testis barrier (BTB) dynamics during spermatogenesis via the "Yin" and "Yang" effects of mammalian target of rapamycin complex 1 (mTORC1) and mTORC2. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:291-358. [PMID: 23317821 DOI: 10.1016/b978-0-12-407704-1.00006-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In mammalian testes, haploid spermatozoa are formed from diploid spermatogonia during spermatogenesis, which is a complicated cellular process. While these cellular events were reported in the 1960s and 1970s, the underlying molecular mechanism(s) that regulates these events remained unexplored until the past ∼10 years. For instance, adhesion proteins were shown to be integrated components at the Sertoli cell-cell interface and/or the Sertoli-spermatid interface in the late 1980s. But only until recently, studies have demonstrated that some of the adhesion proteins serve as the platform for signal transduction that regulates cell adhesion. In this chapter, a brief summary and critical discussion are provided on the latest findings regarding these cell-adhesion proteins in the testis and their relationship to spermatogenesis. Moreover, antagonistic effects of two mammalian target of rapamycin (mTOR) complexes, known as mTORC1 and mTORC2, on cell-adhesion function in the testis are discussed. Finally, a hypothetic model is presented to depict how these two mTOR-signaling complexes having the "yin" and "yang" antagonistic effects on the Sertoli cell tight junction (TJ)-permeability barrier can maintain the blood-testis barrier (BTB) integrity during the epithelial cycle while preleptotene spermatocytes are crossing the BTB.
Collapse
Affiliation(s)
- Ka Wai Mok
- Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, USA
| | | | | |
Collapse
|
86
|
Hexavalent chromium at low concentration alters Sertoli cell barrier and connexin 43 gap junction but not claudin-11 and N-cadherin in the rat seminiferous tubule culture model. Toxicol Appl Pharmacol 2013; 268:27-36. [DOI: 10.1016/j.taap.2013.01.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 11/20/2022]
|
87
|
Abstract
Claudins are tight junction membrane proteins that are expressed in epithelia and endothelia and form paracellular barriers and pores that determine tight junction permeability. This review summarizes our current knowledge of this large protein family and discusses recent advances in our understanding of their structure and physiological functions.
Collapse
Affiliation(s)
- Dorothee Günzel
- Department of Clinical Physiology, Charité, Campus Benjamin Franklin, Berlin, Germany
| | | |
Collapse
|
88
|
Xiao X, Mruk DD, Cheng CY. Intercellular adhesion molecules (ICAMs) and spermatogenesis. Hum Reprod Update 2013; 19:167-86. [PMID: 23287428 DOI: 10.1093/humupd/dms049] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND During the seminiferous epithelial cycle, restructuring takes places at the Sertoli-Sertoli and Sertoli-germ cell interface to accommodate spermatogonia/spermatogonial stem cell renewal via mitosis, cell cycle progression and meiosis, spermiogenesis and spermiation since developing germ cells, in particular spermatids, move 'up and down' the seminiferous epithelium. Furthermore, preleptotene spermatocytes differentiated from type B spermatogonia residing at the basal compartment must traverse the blood-testis barrier (BTB) to enter the adluminal compartment to prepare for meiosis at Stage VIII of the epithelial cycle, a process also accompanied by the release of sperm at spermiation. These cellular events that take place at the opposite ends of the epithelium are co-ordinated by a functional axis designated the apical ectoplasmic specialization (ES)-BTB-basement membrane. However, the regulatory molecules that co-ordinate cellular events in this axis are not known. METHODS Literature was searched at http://www.pubmed.org and http://scholar.google.com to identify published findings regarding intercellular adhesion molecules (ICAMs) and the regulation of this axis. RESULTS Members of the ICAM family, namely ICAM-1 and ICAM-2, and the biologically active soluble ICAM-1 (sICAM-1) are the likely regulatory molecules that co-ordinate these events. sICAM-1 and ICAM-1 have antagonistic effects on the Sertoli cell tight junction-permeability barrier, involved in Sertoli cell BTB restructuring, whereas ICAM-2 is restricted to the apical ES, regulating spermatid adhesion during the epithelial cycle. Studies in other epithelia/endothelia on the role of the ICAM family in regulating cell movement are discussed and this information has been evaluated and integrated into studies of these proteins in the testis to create a hypothetical model, depicting how ICAMs regulate junction restructuring events during spermatogenesis. CONCLUSIONS ICAMs are crucial regulatory molecules of spermatogenesis. The proposed hypothetical model serves as a framework in designing functional experiments for future studies.
Collapse
Affiliation(s)
- Xiang Xiao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
89
|
França LR, Auharek SA, Hess RA, Dufour JM, Hinton BT. Blood-Tissue Barriers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013. [DOI: 10.1007/978-1-4614-4711-5_12] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
90
|
Aponte PM, Schlatt S, Franca LRD. Biotechnological approaches to the treatment of aspermatogenic men. Clinics (Sao Paulo) 2013; 68 Suppl 1:157-67. [PMID: 23503966 PMCID: PMC3583150 DOI: 10.6061/clinics/2013(sup01)18] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 08/30/2012] [Indexed: 01/15/2023] Open
Abstract
Aspermatogenesis is a severe impairment of spermatogenesis in which germ cells are completely lacking or present in an immature form, which results in sterility in approximately 25% of patients. Because assisted reproduction techniques require mature germ cells, biotechnology is a valuable tool for rescuing fertility while maintaining biological fatherhood. However, this process involves, for instance, the differentiation of preexisting immature germ cells or the production/derivation of sperm from somatic cells. This review critically addresses four potential techniques: sperm derivation in vitro, germ stem cell transplantation, xenologous systems, and haploidization. Sperm derivation in vitro is already feasible in fish and mammals through organ culture or 3D systems, and it is very useful in conditions of germ cell arrest or in type II Sertoli-cell-only syndrome. Patients afflicted by type I Sertoli-cell-only syndrome could also benefit from gamete derivation from induced pluripotent stem cells of somatic origin, and human haploid-like cells have already been obtained by using this novel methodology. In the absence of alternative strategies to generate sperm in vitro, in germ cells transplantation fertility is restored by placing donor cells in the recipient germ-cell-free seminiferous epithelium, which has proven effective in conditions of spermatogonial arrest. Grafting also provides an approach for ex-vivo generation of mature sperm, particularly using prepubertal testis tissue. Although less feasible, haploidization is an option for creating gametes based on biological cloning technology. In conclusion, the aforementioned promising techniques remain largely experimental and still require extensive research, which should address, among other concerns, ethical and biosafety issues, such as gamete epigenetic status, ploidy, and chromatin integrity.
Collapse
Affiliation(s)
- Pedro Manuel Aponte
- Department of Morphology, Federal University of Minas Gerais, Minas Gerais, Brazil
| | | | | |
Collapse
|
91
|
Pérez CV, Sobarzo CM, Jacobo PV, Pellizzari EH, Cigorraga SB, Denduchis B, Lustig L. Loss of occludin expression and impairment of blood-testis barrier permeability in rats with autoimmune orchitis: effect of interleukin 6 on Sertoli cell tight junctions. Biol Reprod 2012; 87:122. [PMID: 23018187 DOI: 10.1095/biolreprod.112.101709] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Inflammation of the male reproductive tract is accepted as being an important etiological factor of infertility. Experimental autoimmune orchitis (EAO) is characterized by interstitial lymphomononuclear cell infiltration and severe damage of seminiferous tubules with germ cells that undergo apoptosis and sloughing. Because the blood-testis barrier (BTB) is relevant for the protection of haploid germ cells against immune attack, the aim of this study was to analyze BTB permeability and the expression of tight junction proteins (occludin, claudin 11, and tight junction protein 1 [TJP1]) in rats during development of autoimmune orchitis. The role of IL6 as modulator of tight junction dynamics was also evaluated because intratesticular content of this cytokine is increased in EAO rats. Orchitis was induced in Sprague-Dawley adult rats by active immunization with testicular homogenate and adjuvants. Control rats (C) were injected with saline solution and adjuvants. Untreated (N) rats were also studied. Concomitant with early signs of germ cell sloughing, a reduced expression of occludin and delocalization of claudin 11 and TJP1 were detected in the testes of rats with EAO compared to C and N groups. The use of tracers showed increased BTB permeability in EAO rats. Intratesticular injection of IL6 induced focal testicular inflammation, which is associated with damaged seminiferous tubules. Rat Sertoli cells cultured in the presence of IL6 exhibited a redistribution of tight junction proteins and reduced transepithelial electrical resistance. These data indicate the possibility that IL6 might be involved in the downregulation of occludin expression and in the modulation of BTB permeability that occur in rats undergoing autoimmune orchitis.
Collapse
Affiliation(s)
- Cecilia Valeria Pérez
- Instituto de Investigaciones Biomédicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
92
|
Expression Pattern of Testicular claudin-11 in Infertile Men. Urology 2012; 80:1161.e13-7. [DOI: 10.1016/j.urology.2012.06.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 05/09/2012] [Accepted: 06/26/2012] [Indexed: 11/21/2022]
|
93
|
Bhattacharya I, Pradhan BS, Sarda K, Gautam M, Basu S, Majumdar SS. A switch in Sertoli cell responsiveness to FSH may be responsible for robust onset of germ cell differentiation during prepubartal testicular maturation in rats. Am J Physiol Endocrinol Metab 2012; 303:E886-98. [PMID: 22850685 DOI: 10.1152/ajpendo.00293.2012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
FSH and Testosterone (T) regulate spermatogenesis via testicular Sertoli cells (Sc), which bear receptors for these hormones. Despite sufficient circulating levels of FSH and T postnatally, predominant appearance of spermatogonia B and spermatocytes is not discernible until 11 and 18 days of postnatal age, respectively, in rat testes. In an attempt to explore the underlying causes, we cultured Sc from neonatal (5- and 9-day-old) and prepubertal (12- and 19-day-old) rat testes and compared the status of FSH receptor (FSH-R) and androgen receptor (AR) signaling. Protein and mRNA levels of FSH-R and AR remained uniform in cultured Sc from all age groups. Androgen binding ability of AR was similar, and T-induced nuclear localization of AR was discernible in Sc from all age groups. Binding of FSH to FSH-R, subsequent production of cAMP, and mRNA of stem cell factor (SCF) and glial cell line-derived neurotrophic factor (GDNF), known to be essential for the robust differentiation of repopulating spermatogonia, were significantly augmented in prepubertal Sc compared with those in neonatal Sc. However, treatment of neonatal Sc with cholera toxin or forskolin, which stimulate cAMP production bypassing FSH-R, demonstrated a concomitant rise in SCF and GDNF mRNA expression, which was similar to the FSH-mediated rise observed in prepubertal Sc. These observations suggested that, during prepubertal Sc maturation, the ability of FSH-R to respond to FSH is significantly augmented and is associated with the robust differentiation of repopulating spermatogonia, and such a switch in Sc from FSH-resistant to FSH-responsive mode during prepubertal development may underlie the initiation of robust spermatogenesis.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Cellular Endocrinology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, JNU Complex, New Delhi 110067, India
| | | | | | | | | | | |
Collapse
|
94
|
Majumdar SS, Sarda K, Bhattacharya I, Plant TM. Insufficient androgen and FSH signaling may be responsible for the azoospermia of the infantile primate testes despite exposure to an adult-like hormonal milieu. Hum Reprod 2012; 27:2515-25. [PMID: 22669085 DOI: 10.1093/humrep/des184] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND In humans, as well as in other higher primates, the infantile testis is exposed to an adult-like hormonal milieu, but spermatogenesis is not initiated at this stage of primate development. In the present study, we examined the molecular basis of this intriguing infertile state of the primate testis. METHODS The integrity of androgen receptor (AR) and FSH receptor (FSHR) signaling pathways in primary cultures of Sertoli cells (Scs) harvested from azoospermic infant and spermatogenic pubertal monkey testes were investigated under identical in vitro hormonal conditions. In order to synchronously harvest Scs from early pubertal testis, the activation of testicular puberty was timed experimentally by prematurely initiating gonadotrophin secretion in juvenile animals with an intermittent infusion of gonadotrophin-releasing hormone. RESULTS While qRT-PCR demonstrated that AR and FSHR mRNA expression in Scs from infant and pubertal testes were comparable, androgen-binding and FSH-mediated cAMP production by infant Scs was extremely low. Compromised AR and FSHR signaling in infant Scs was further supported by the finding that testosterone (T) and FSH failed to augment the expression of the T responsive gene, claudin 11, and the FSH responsive genes, inhibin-βB, stem cell factor (SCF) and glial cell line-derived neurotrophic factor (GDNF) in Scs harvested at this stage of development. CONCLUSION These results indicate that compromised AR and FSHR signaling pathways in Scs underlie the inability of the infant primate testis to respond to an endogenous hormonal milieu that later in development, at the time puberty, stimulates the initiation of spermatogenesis. This finding may have relevance to some forms of idiopathic infertility in men.
Collapse
Affiliation(s)
- Subeer S Majumdar
- Division of Cellular Endocrinology, National Institute of Immunology, New Delhi 110067, India.
| | | | | | | |
Collapse
|
95
|
Abstract
The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. It divides the seminiferous epithelium into the basal and the apical (adluminal) compartments. Meiosis I and II, spermiogenesis, and spermiation all take place in a specialized microenvironment behind the BTB in the apical compartment, but spermatogonial renewal and differentiation and cell cycle progression up to the preleptotene spermatocyte stage take place outside of the BTB in the basal compartment of the epithelium. However, the BTB is not a static ultrastructure. Instead, it undergoes extensive restructuring during the seminiferous epithelial cycle of spermatogenesis at stage VIII to allow the transit of preleptotene spermatocytes at the BTB. Yet the immunological barrier conferred by the BTB cannot be compromised, even transiently, during the epithelial cycle to avoid the production of antibodies against meiotic and postmeiotic germ cells. Studies have demonstrated that some unlikely partners, namely adhesion protein complexes (e.g., occludin-ZO-1, N-cadherin-β-catenin, claudin-5-ZO-1), steroids (e.g., testosterone, estradiol-17β), nonreceptor protein kinases (e.g., focal adhesion kinase, c-Src, c-Yes), polarity proteins (e.g., PAR6, Cdc42, 14-3-3), endocytic vesicle proteins (e.g., clathrin, caveolin, dynamin 2), and actin regulatory proteins (e.g., Eps8, Arp2/3 complex), are working together, apparently under the overall influence of cytokines (e.g., transforming growth factor-β3, tumor necrosis factor-α, interleukin-1α). In short, a "new" BTB is created behind spermatocytes in transit while the "old" BTB above transiting cells undergoes timely degeneration, so that the immunological barrier can be maintained while spermatocytes are traversing the BTB. We also discuss recent findings regarding the molecular mechanisms by which environmental toxicants (e.g., cadmium, bisphenol A) induce testicular injury via their initial actions at the BTB to elicit subsequent damage to germ-cell adhesion, thereby leading to germ-cell loss, reduced sperm count, and male infertility or subfertility. Moreover, we also critically evaluate findings in the field regarding studies on drug transporters in the testis and discuss how these influx and efflux pumps regulate the entry of potential nonhormonal male contraceptives to the apical compartment to exert their effects. Collectively, these findings illustrate multiple potential targets are present at the BTB for innovative contraceptive development and for better delivery of drugs to alleviate toxicant-induced reproductive dysfunction in men.
Collapse
Affiliation(s)
- C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA.
| | | |
Collapse
|
96
|
Tanwar PS, Zhang L, Teixeira JM. Adenomatous polyposis coli (APC) is essential for maintaining the integrity of the seminiferous epithelium. Mol Endocrinol 2011; 25:1725-39. [PMID: 21816903 DOI: 10.1210/me.2011-0057] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sertoli cells provide the microenvironment necessary for germ cell development and spermatogenesis; disruption of Sertoli cell morphology or function can lead to germ cell aplasia, which is observed in testicular dysgenesis syndrome. Mutation of the adenomatous polyposis coli (APC) gene has been associated with various human cancers, including testicular cancer, but its involvement in nonmalignant testicular pathologies has not been reported. We have developed a mouse model (APC(cko)) that expresses a truncated form of APC in Sertoli cells. Despite normal embryonic and early postnatal testicular development in APC(cko) mice, premature germ cell loss and Sertoli cell-only seminiferous tubules were observed in mutant testes without affecting Sertoli cell quiescence, apoptosis, or differentiation, which were confirmed by the absence of both proliferating cell nuclear antigen, DNA strand breaks, and anti-Müllerian hormone, respectively. We show that mutant Sertoli cells lose their apical extensions, which would normally enclose germ cells during various stages of spermatogenesis, and were unable to maintain the blood-testis barrier because of disrupted expression of junctional proteins. We also observed an up-regulation of Snail and Slug, markers suggestive of epithelial-mesenchymal transition in the Sertoli cells, but tumorigenesis was not observed. No comparable phenotype was observed with Sertoli cell-specific loss-of-function mutations in β-catenin, leading us to speculate that truncation of APC in Sertoli cells results in progressive degeneration of the seminiferous tubules by a mechanism that disrupts the integrity of Sertoli cell junctions independently of APC-regulated β-catenin activities and leads to development of a Sertoli cell-only phenotype.
Collapse
Affiliation(s)
- Pradeep S Tanwar
- Vincent Center for Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
97
|
Abstract
The blood-testis barrier (BTB), which is created by adjacent Sertoli cells near the basement membrane, serves as a 'gatekeeper' to prohibit harmful substances from reaching developing germ cells, most notably postmeiotic spermatids. The BTB also divides the seminiferous epithelium into the basal and adluminal (apical) compartment so that postmeiotic spermatid development, namely spermiogenesis, can take place in a specialized microenvironment in the apical compartment behind the BTB. The BTB also contributes, at least in part, to the immune privilege status of the testis, so that anti-sperm antibodies are not developed against antigens that are expressed transiently during spermatogenesis. Recent studies have shown that numerous drug transporters are expressed by Sertoli cells. However, many of these same drug transporters are also expressed by spermatogonia, spermatocytes, round spermatids, elongating spermatids, and elongated spermatids, suggesting that the developing germ cells are also able to selectively pump drugs 'in' and/or 'out' via influx or efflux pumps. We review herein the latest developments regarding the role of drug transporters in spermatogenesis. We also propose a model utilized by the testis to protect germ cell development from 'harmful' environmental toxicants and xenobiotics and/or from 'therapeutic' substances (e.g. anticancer drugs). We also discuss how drug transporters that are supposed to protect spermatogenesis can work against the testis in some instances. For example, when drugs (e.g. male contraceptives) that can perturb germ cell adhesion and/or maturation are actively pumped out of the testis or are prevented from entering the apical compartment, such as by efflux pumps.
Collapse
Affiliation(s)
- Linlin Su
- The Mary M Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York 10065, USA
| | | | | |
Collapse
|
98
|
Park C, Lee J, Oh Y, Shim S, Nah W, Choi K, Gye M. Expression of claudin-1 and -11 in immature and mature pheasant (Phasianus colchicus) testes. Theriogenology 2011; 75:445-58. [DOI: 10.1016/j.theriogenology.2010.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 09/06/2010] [Accepted: 09/10/2010] [Indexed: 11/27/2022]
|
99
|
Cheng CY, Mruk DD. The biology of spermatogenesis: the past, present and future. Philos Trans R Soc Lond B Biol Sci 2010; 365:1459-63. [PMID: 20403863 DOI: 10.1098/rstb.2010.0024] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The physiological function of spermatogenesis in Caenorhabditis elegans, Drosophila melanogaster and mammals is to produce spermatozoa (1n, haploid) that contain only half of the genetic material of spermatogonia (2n, diploid). This half number of chromosomes from a spermatozoon will then be reconstituted to become a diploid cell upon fertilization with an egg, which is also haploid. Thus, genetic information from two parental individuals can be passed onto their offspring. Spermatogenesis takes place in the seminiferous epithelium of the seminiferous tubule, the functional unit of the mammalian testis. In mammals, particularly in rodents, the fascinating morphological changes that occur during spermatogenesis involving cellular differentiation and transformation, mitosis, meiosis, germ cell movement, spermiogenesis and spermiation have been well documented from the 1950s through the 1980s. During this time, however, the regulation of, as well as the biochemical and molecular mechanisms underlying these diverse cellular events occurring throughout spermatogenesis, have remained largely unexplored. In the past two decades, important advancements have been made using new biochemical, cell and molecular biology techniques to understand how different genes, proteins and signalling pathways regulate various aspects of spermatogenesis. These include studies on the differentiation of spermatogonia from gonocytes; regulation of spermatogonial stem cells; regulation of spermatogonial mitosis; regulation of meiosis, spermiogenesis and spermiation; role of hormones (e.g. oestrogens, androgens) in spermatogenesis; transcriptional regulation of spermatogenesis; regulation of apoptosis; cell-cell interactions; and the biology of junction dynamics during spermatogenesis. The impact of environmental toxicants on spermatogenesis has also become an urgent issue in the field in light of declining fertility levels in males. Many of these studies have helped investigators to understand important similarities, differences and evolutionary relationships between C. elegans, D. melanogaster and mammals relating to spermatogenesis. In this Special Issue of the Philosophical Transactions of the Royal Society B: Biological Sciences, we have covered many of these areas, and in this Introduction, we highlight the topic of spermatogenesis by examining its past, present and future.
Collapse
Affiliation(s)
- C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Population Council, 1230 York Avenue, New York, NY 10065, USA.
| | | |
Collapse
|