Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Reproduction ; 156(1): 35-46, 2018 07.
Article in English | MEDLINE | ID: mdl-29743262

ABSTRACT

The testis is an organ that maintains an immune suppressive environment. We previously revealed that exposure of pre-pubertal rats to an acute dose of a well-described Sertoli cell toxicant, mono-(2-ethylhexyl) phthalate (MEHP), leads to an accumulation of CD11b+ immune cells in the testicular interstitial space that closely correlates with a robust incidence of germ cell (GC) apoptosis. Here, we test the hypothesis that the infiltrating immune cells contribute to GC apoptosis. Postnatal day 28 Fischer rats that received an oral dose of 700 mg/kg MEHP showed a significant infiltration of both CD11bc+/CD68+/CD163- macrophages and neutrophils. The infiltration peaked at 12 h, but had reduced by 48 h. Testicular macrophages from MEHP-treated rats showed significantly upregulated expression of Tnfa and Il6, and the Arg1/Nos2 ratio was reduced compared to controls. However, small increases in anti-inflammatory genes Il10 and Tgfb1 were also observed. Depletion of circulating monocytes with clodronate liposomes prior to MEHP treatment reduced the macrophage influx into the testis, but did not lower GC apoptosis. Additionally, depletion of neutrophils using an anti-polymorphonuclear cell antibody prevented both macrophage and neutrophil infiltration into the testis, and also did not affect GC apoptosis. Together, these results show that exposure to MEHP leads to a rapid and temporary influx of pro-inflammatory monocytes and neutrophils in the interstitium of the testis. However, with this acute dosing paradigm, these infiltrating leukocytes do not appear to contribute to MEHP-induced testicular GC apoptosis leaving the functional significance of these infiltrating cells in the pathogenesis of MEHP-induced testicular injury unresolved.


Subject(s)
Apoptosis/drug effects , Diethylhexyl Phthalate/analogs & derivatives , Orchitis/pathology , Spermatozoa/drug effects , Testis/drug effects , Animals , Diethylhexyl Phthalate/pharmacology , Macrophages/drug effects , Macrophages/pathology , Male , Rats , Spermatozoa/pathology , Testis/pathology
2.
Semin Cell Dev Biol ; 30: 27-35, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24632385

ABSTRACT

The ubiquitination of proteins is a post-translational modification that was first described as a means to target misfolded or unwanted proteins for degradation by the proteasome. It is now appreciated that the ubiquitination of proteins also serves as a mechanism to modify protein function and cellular functions such as protein trafficking, cell signaling, DNA repair, chromatin modifications, cell-cycle progression and cell death. The ubiquitination of proteins occurs through the hierarchal transfer of ubiquitin from an E1 ubiquitin-activating enzyme to an E2 ubiquitin-conjugating enzyme and finally to an E3 ubiquitin ligase that transfers the ubiquitin to its target protein. It is the final E3 ubiquitin ligase that confers the substrate specificity for ubiquitination and is the focus of this review. Spermatogenesis is a complex and highly regulated process by which spermatogonial stem cells undergo mitotic proliferation and expansion of the diploid spermatogonial population, differentiate into spermatocytes and progress through two meiotic divisions to produce haploid spermatids that proceed through a final morphogenesis to generate mature spermatozoa. The ubiquitination of proteins in the cells of the testis occurs in many of the processes required for the progression of mature spermatozoa. Since it is the E3 ubiquitin ligase that recognizes the target protein and provides the specificity and selectivity for ubiquitination, this review highlights known examples of E3 ligases in the testis and the differing roles that they play in maintaining functional spermatogenesis.


Subject(s)
Spermatogenesis , Ubiquitin-Protein Ligases/physiology , Ubiquitination , Animals , Humans , Intercellular Junctions/metabolism , Male , Proteolysis , Signal Transduction , Testis/cytology , Testis/physiology
3.
Biol Reprod ; 91(1): 18, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24876407

ABSTRACT

The mechanism by which noninfectious testicular inflammation results in infertility is poorly understood. Here the infiltration of CD11b+ immunoreactive testicular interstitial cells (neutrophil, macrophages, dendritic cells) in immature (Postnatal Day [PND] 21, 28, and 35) and adult (PND 56) Fischer rats is described at 12, 24, and 48 h after an oral dose of 1 g/kg mono-(2-ethylhexyl) phthalate (MEHP), a well-described Sertoli cell toxicant. Increases of CD11b+ cells are evident 12 h after MEHP exposure in PND 21 and 28 rats. In PND 28 rats, CD11b+ cells remained significantly elevated at 48 h, while in PND 21 rats, it returned to control levels by 24 h. The peak number of CD11b+ cells in PND 35 rat testis is delayed until 24 h, but remains significantly elevated at 48 h. In PND 56 rats, no increase in CD11b+ cells occurs after MEHP exposure. In PND 21, 28, and 35 rats, a significant increase in monocyte chemoattractant protein-1 (MCP-1) by peritubular myoid cells occurs 12 h after MEHP. Interestingly, MEHP treatment of C57BL/6J mice did not incite an infiltration of CD11b+ cells at either PND 21 or 28. The peak level of germ cell apoptosis observed 24 h after MEHP exposure in young rats is not seen in mice at any age or in PND 56 rats. Taken together, these findings implicate MCP-1 released by peritubular myoid cells in provoking the migration of CD11b+ cells into the immature rat testis early after MEHP exposure and point to a role for CD11b+ cells in triggering germ cell apoptosis in an age- and species-dependent manner.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Macrophages/drug effects , Sertoli Cells/drug effects , Testis/drug effects , Age Factors , Animals , Cell Movement/physiology , Chemokine CCL2/metabolism , Diethylhexyl Phthalate/pharmacology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Rats , Sertoli Cells/immunology , Sertoli Cells/metabolism , Species Specificity , Testis/immunology , Testis/metabolism
4.
Toxicol Sci ; 200(1): 70-78, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38565259

ABSTRACT

Peritubular macrophages (PTMφ) are predominantly localized near spermatogonial stem cells in the testis. We previously revealed that exposure of peripubertal male Fischer rats to mono-(2-ethylhexyl) phthalate (MEHP) leads to increased PTMφs in the testis. The mechanisms that trigger increases in PTMφs in the testis are poorly understood. However, MEHP exposure is known to both induce spermatocyte apoptosis and to perturb the blood-testis barrier (BTB). This study aims to elucidate the association between the disruption of BTB and the increases of PTMφs in the testis by comparing the effects observed with MEHP to 2 other testicular toxicants with variable effects on the BTB and subtype of germ cell undergoing apoptosis. Methoxyacetic acid (MAA) acts directly on spermatocytes and does not affect BTB function, whereas cadmium chloride (CdCl2) induces profound injury to BTB. The results indicated that MAA exposure significantly increased spermatocyte apoptosis, whereas no significant changes in the numbers of PTMφs in the testis occurred. In contrast, CdCl2 exposure disrupted BTB function and increased the abundance of PTMφs in the testis. To further investigate whether MEHP-induced changes in BTB integrity accounted for the increase in PTMφs, a plasmid for LG3/4/5, the functional component of laminin-alpha 2, was overexpressed in the testis to stabilize BTB integrity before MEHP exposure. The results showed that LG3/4/5 overexpression substantially reduced the ability of MEHP to compromise BTB integrity and prevented the increase in PTMφ numbers after MEHP exposure. These results indicate that BTB disruption is necessary to increase PTMφs in the testis induced by toxicants.


Subject(s)
Apoptosis , Blood-Testis Barrier , Diethylhexyl Phthalate , Macrophages , Rats, Inbred F344 , Testis , Animals , Male , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/pathology , Blood-Testis Barrier/metabolism , Diethylhexyl Phthalate/toxicity , Diethylhexyl Phthalate/analogs & derivatives , Testis/drug effects , Testis/pathology , Testis/metabolism , Macrophages/drug effects , Apoptosis/drug effects , Cadmium Chloride/toxicity , Acetates/toxicity , Rats , Spermatocytes/drug effects , Spermatocytes/pathology
5.
Toxicol Sci ; 198(1): 76-85, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38113427

ABSTRACT

Exposure of rodents to mono-(2-ethylhexyl) phthalate (MEHP) is known to disrupt the blood-testis barrier and cause testicular germ cell apoptosis. Peritubular macrophages (PTMφ) are a newly identified type of testicular macrophage that aggregates near the spermatogonial stem cell niche. We have previously reported that MEHP exposure increased the numbers of PTMφs by 6-fold within the testis of peripubertal rats. The underlying mechanism(s) accounting for this change in PTMφs and its biological significance is unknown. This study investigates if MEHP-induced alterations in PTMφs occur in rodents (PND 75 adult rats and PND 26 peripubertal mice) that are known to be less sensitive to MEHP-induced testicular toxicity. Results show that adult rats have a 2-fold higher basal level of PTMφ numbers than species-matched peripubertal animals, but there was no significant increase in PTMφ numbers after MEHP exposure. Peripubertal mice have a 5-fold higher basal level of PTMφ compared with peripubertal rats but did not exhibit increases in number after MEHP exposure. Further, the interrogation of the testis transcriptome was profiled from both the MEHP-responsive peripubertal rats and the less sensitive rodents via 3' Tag sequencing. Significant changes in gene expression were observed in peripubertal rats after MEHP exposure. However, adult rats showed lesser changes in gene expression, and peripubertal mice showed only minor changes. Collectively, the data show that PTMφ numbers are associated with the sensitivity of rodents to MEHP in an age- and species-dependent manner.


Subject(s)
Diethylhexyl Phthalate , Diethylhexyl Phthalate/analogs & derivatives , Testis , Male , Rats , Mice , Animals , Transcriptome , Sertoli Cells , Rodentia , Diethylhexyl Phthalate/toxicity , Macrophages
6.
Toxicol Sci ; 2023 Nov 06.
Article in English | MEDLINE | ID: mdl-37941498

ABSTRACT

The blood-testis barrier (BTB) is constituted by tight junctions between adjacent Sertoli cells (SC) that create a specialized adluminal microenvironment to foster the development of spermatocytes and spermatids. The BTB is a well-studied target of numerous environmental toxicants, including di-(2-ethylhexyl) phthalate (DEHP), a compound widely used in various consumer products. MEHP is the active toxic metabolite of DEHP that has long been recognized in postnatal rodents to disrupt SC function. This study evaluates the impact of MEHP on the integrity of the BTB in both pubertal and adult rats and the signal transduction pathways known to be involved in the disruption of the BTB. Treatment of prepubertal rats with 700 mg/kg MEHP for 24 hours functionally disrupted the BTB integrity. A similar treatment of adult rats with MEHP did not disrupt the integrity of the BTB. The observed disruption of the BTB integrity in the MEHP-treated prepubertal rats occurred concomitantly with a decreased expression and mislocalization of both the ZO1 and occludin tight junction-associated proteins, as well as sloughing of spermatocytes and spermatids. At this same time, MEHP treatment induced a transient surge of p44/42 mitogen-activated protein kinase (MAPK) pathway. Interestingly, after a recovery period of 5 weeks, the BTB recovered and was functionally intact. This is the first report to indicate that acute MEHP exposure of prepubertal rats, but not adult rats, disrupts the functional integrity of the BTB and that this effect on the BTB is reversible.

7.
Biol Reprod ; 86(5): 160, 1-10, 2012 May.
Article in English | MEDLINE | ID: mdl-22321834

ABSTRACT

Testicular dysgenesis syndrome refers to a collection of diseases in men, including testicular cancer, that arise as a result of abnormal testicular development. Phthalates are a class of chemicals used widely in the production of plastic products and other consumer goods. Unfortunately, phthalate exposure has been linked to reproductive dysfunction and has been shown to adversely affect normal germ cell development. In this study, we show that mono-(2-ethylhexyl) phthalate (MEHP) induces matrix metalloproteinase 2 (MMP2) expression in testicular embryonal carcinoma NT2/D1 cells but has no significant effect on MMP9 expression. NT2/D1 cells also have higher levels of MYC expression following MEHP treatment. It is widely recognized that activation of MMP2 and MYC is tightly associated with tumor metastasis and tumor progression. Gelatin zymographic analysis indicates that MEHP strongly activates MMP2 in NT2/D1 cells. Addition of the MMP2-specific inhibitor SB-3CT inhibited MEHP-enhanced cell invasion and migration, demonstrating that MMP2 plays a functional role in promoting testicular embryonal carcinoma progression in response to MEHP exposure. Furthermore, we investigated genome-wide gene expression profiles of NT2/D1 cells following MEHP exposure at 0, 3, and 24 h. Microarray analysis and semiquantitative RT-PCR revealed that MEHP exposure primarily influenced genes in cell adhesion and transcription in NT2/D1 cells. Gap junction protein-alpha 1, vinculin, and inhibitor of DNA-binding protein-1 were significantly down-regulated by MEHP treatment, while claudin-6 and beta 1-catenin expression levels were up-regulated. This study provides insight into mechanisms that may account for modulating testicular cancer progression following phthalate exposure.


Subject(s)
Cell Movement/physiology , Diethylhexyl Phthalate/analogs & derivatives , Embryonal Carcinoma Stem Cells/pathology , Testicular Neoplasms/pathology , Catenins/biosynthesis , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/drug effects , Claudins/biosynthesis , Connexin 43/biosynthesis , Diethylhexyl Phthalate/adverse effects , Embryonal Carcinoma Stem Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Heterocyclic Compounds, 1-Ring/pharmacology , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Neoplasm Invasiveness , Sulfones/pharmacology , Testicular Neoplasms/drug therapy , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Vinculin/biosynthesis , Delta Catenin
8.
Biol Reprod ; 85(6): 1203-15, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21832167

ABSTRACT

Our previous studies showed that the prototypical testicular toxic phthalate monoester, mono-(2-ethylhexyl) phthalate (MEHP), suppresses Sertoli cell TIMP2 levels and allows for the activation of MMP2 in seminiferous epithelium. Activation of MMP2 is important for triggering germ cell apoptosis and instigating germ cell detachment from Sertoli cells. These novel findings led us to examine the transcriptional regulation of the Timp2 gene that accounts for the decrease in Sertoli cell TIMP2 levels following MEHP exposure. Sequential deletion of the Timp2 5'-upstream activating sequence (1200 bp) was used to survey transcriptional activation in the Timp2 promoter region in response to MEHP. Results indicate that under control conditions in rat Sertoli cells, CCAAT enhancer-binding protein alpha (CEBPA) acts as a transactivator to initiate Timp2 gene transcription, and its action is deactivated by exposure to MEHP. By contrast, MYC protein acts as an inhibitor of Timp2 gene transcription, and its activity is increased after MEHP treatment. Addition of follicle-stimulating hormone (FSH) to cells causes translocation of CEBPA into the Sertoli cell nucleus and rescues MEHP-suppressed TIMP2 levels. Down-regulation of TIMP2 expression by MEHP exposure is blocked by forskolin (a cAMP-elevating agent), suggesting that the decrease in Sertoli cell TIMP2 expression following MEHP exposure is cAMP-dependent. Taken together, these data indicate that MEHP both disrupts the FSH-stimulated cAMP signaling pathway and activates the inhibitory signaling mediated by MYC protein, to ultimately account for the cellular mechanism underlying the decreased expression of TIMP2 in Sertoli cells.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Diethylhexyl Phthalate/analogs & derivatives , Proto-Oncogene Proteins c-myc/metabolism , Sertoli Cells/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Animals , Coculture Techniques , Diethylhexyl Phthalate/toxicity , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , RNA, Messenger/metabolism , Second Messenger Systems , Sertoli Cells/drug effects , Tissue Inhibitor of Metalloproteinase-2/genetics
9.
Toxicol Sci ; 182(2): 288-296, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34010400

ABSTRACT

Peripubertal exposure of male rodents to the phthalate metabolite mono-(2-ethylhexyl) phthalate (MEHP) causes testicular inflammation, spermatocyte apoptosis, and disruption of the blood-testis barrier. The MEHP-induced inflammatory response in the testis includes an infiltration of macrophages and neutrophils, although the cause and purpose of this response is unknown. Recently, a population of testicular macrophages known as peritubular macrophages that are phenotypically distinct from those resident in interstitium was described in mice. Peritubular macrophages aggregate near the spermatogonial stem cell niche and are believed to stimulate their differentiation. We hypothesized that if testicular peritubular macrophages do indeed stimulate spermatogonial differentiation, MEHP exposure would result in an increase of peritubular macrophages to stimulate the replacement of lost spermatocytes. Male rats were exposed to 700 mg/kg MEHP or corn oil (vehicle control) via oral gavage at postnatal day 28 and euthanized at 48 h, 1 or 2 weeks later. Seminiferous tubules were stained with immunofluorescent markers for macrophages (major histocompatibility complex class II [MHC-II+]) and undifferentiated spermatogonia (PLZF). Peritubular macrophages were observed in rat testis: MHC-II+ cells on the surface of seminiferous tubules with heterogeneous morphology. Quantification of MHC-II+ cells revealed that, unlike in the mouse, their numbers did not increase through puberty (2-week period). MEHP increased macrophage presence by 6-fold 48 h after exposure and remained elevated by 2-fold 2 weeks after exposure. An increase of differentiating spermatogonia occurred 2 weeks after MEHP exposure. Taken together, our results suggest that peritubular macrophages play a crucial role in the testis response to acute injury and the subsequent recovery of spermatogenesis.


Subject(s)
Diethylhexyl Phthalate , Testis , Animals , Diethylhexyl Phthalate/analogs & derivatives , Diethylhexyl Phthalate/toxicity , Macrophages , Male , Mice , Phthalic Acids , Rats , Spermatogonia
10.
Biol Reprod ; 82(3): 516-27, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19828778

ABSTRACT

Tight junctions between Sertoli cells of the testicular seminiferous epithelium establishes the blood-testis barrier (BTB) and creates a specialized adluminal microenvironment above the BTB that is required for the development of the germ cells that reside there. Actin filament-based anchoring junctions between Sertoli cells and germ cells are important for maintaining close physical contact between these cells as well as regulating the release of mature spermatids into the lumen. Previously, we reported that Sertoli cell injury in rodents after mono-(2-ethylhexyl) phthalate (MEHP) exposure results in the activation of matrix metalloproteinase 2 (MMP2) and increases the sensitivity of germ cells to undergo apoptosis. A disruption in the physical association between Sertoli cells and germ cells and premature loss of germ cells from the seminiferous epithelium has been widely described after phthalate treatment. In this study, we investigate the functional participation of MMP2 in the mechanism underlying MEHP-induced disruption of junction complexes and the resultant loss of germ cells. Exposure of C57BL/6J mice to MEHP (1 g/kg, oral gavage) decreased the expression of occludin in the tight junctions between Sertoli cells and caused gaps between adjacent Sertoli cells as observed by transmission electron microscopy. A reduced expression of laminin-gamma3 and beta1-integrin in apical ectoplasmic specializations between Sertoli cells and germ cells in a time-dependent manner was also observed. Treatment with specific MMP2 inhibitors (TIMP2 and SB-3CT) both in vitro and in vivo significantly suppressed MEHP-induced germ cell sloughing and changes in the expression of these junctional proteins, indicating that MMP-2 plays a primary role in this process. Furthermore, the detachment of germ cells from Sertoli cells appears to be independent of the apoptotic signaling process since MEHP-induced germ cell detachment from Sertoli cells could not be prevented by the addition of a pan-caspase inhibitor (z-VAD-FMK).


Subject(s)
Adherens Junctions/drug effects , Diethylhexyl Phthalate/analogs & derivatives , Matrix Metalloproteinase 2/physiology , Seminiferous Epithelium/drug effects , Testis/drug effects , Adherens Junctions/metabolism , Animals , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Diethylhexyl Phthalate/pharmacology , Enzyme Activation/drug effects , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Multiprotein Complexes/drug effects , Multiprotein Complexes/metabolism , Rats , Rats, Inbred F344 , Seminiferous Epithelium/metabolism , Testicular Diseases/chemically induced , Testicular Diseases/metabolism , Testis/metabolism , Testis/ultrastructure
11.
Toxicol Res (Camb) ; 8(6): 972-978, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-32665841

ABSTRACT

Exposure to the chemotherapeutic agent cis-diamminedichloroplatinum(ii) (cDDP) is well known to instigate acute and prolonged testicular injury in male patients. Many investigators have hypothesized that cDDP-induced dysfunction of Sertoli cells (SCs) may, in part, account for the cDDP-induced lasting testicular injury. Nevertheless, the relative contribution of cDDP-induced SC injury versus direct effects on germ cells (GCs) to the pathogenesis of GC loss remains to be elucidated. The expression of the copper transporter 1 (CTR1) protein in cells directly corresponds with cDDP uptake and its cellular toxicity. Therefore, to discern the role of SCs in the pathogenic mechanism, mice were developed with a SC-specific disruption of the Ctr1 gene (SC ΔCtr1 ) as a strategy to prevent their exposure to cDDP. Adult mice at postnatal day (PND) 60 were treated with 5 mg kg-1 cDDP and then testis collected at 48 hours. A two-fold increase in GC-apoptosis occurred in the testis of cDDP-treated wildtype (WT) mice as compared to saline-treated WT mice. In contrast, cDDP-treated SC ΔCtr1 mice exhibited only a half-fold increase in GC-apoptosis as compared to the saline-treated SC ΔCtr1 mice. This reduced incidence of GC apoptosis in the SC ΔCtr1 mice corresponded to a significantly lower level of platinum within the testis. Taken together, these findings reveal that the uptake of cDDP by CTR1 in SCs accounts for the accumulation of cDDP in the testis and plays a pivotal role in the pathogenic sequence of events leading to the loss of germ cells via apoptosis.

12.
PLoS One ; 14(4): e0215522, 2019.
Article in English | MEDLINE | ID: mdl-31002737

ABSTRACT

An imbalance in copper (Cu) tissue homeostasis has a degenerative effect on spermatogenesis and male fertility. The high-affinity Cu transporter 1 (CTR1; SLC31A1) is the major protein responsible for Cu acquisition in eukaryotes and is highly expressed in mouse testes. Studies on yeast and Drosophila have demonstrated the conserved essential function of Cu and CTR1 for meiosis and fertility, implying that CTR1 may play an essential function in mammalian spermatogenesis. In mice, spermatogenesis takes place within the seminiferous epithelium, where tight junctions between somatic Sertoli cells (SCs) create a specialized microenvironment for the development of meiotic germ cells (GCs) by tightly regulating the free transport of metabolites and ions to reach these cells. Here, it is demonstrated that within the seminiferous epithelium, CTR1 is expressed on the membrane of primary pachytene spermatocytes and SCs. To examine the physiological significance of CTR1 in spermatogenesis, mice with a GC-specific (Ctr1ΔGC) and SC-specific (Ctr1ΔSC) disruption of the Ctr1 gene were generated. The testis of Ctr1ΔGC mice exhibits a severe progressive loss of GCs starting at postnatal day (PND) 28 leading to testis hypoplasia by adulthood. No spermatogenic recovery was observed in Ctr1ΔGC testis beyond PND 41, despite the presence of FOXO-1 expressing undifferentiated spermatogonial cells. However, Ctr1ΔSC mice displayed functional spermatogenesis and were fertile, even though testicular Cu levels and Cu-dependent cellular activities were significantly reduced. These results reveal, for the first time, the importance of CTR1 expression by GCs for maintaining functional spermatogenesis.


Subject(s)
Copper Transporter 1/genetics , Gene Expression , Sertoli Cells/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Testis/metabolism , Animals , Copper/metabolism , Copper Transporter 1/metabolism , Fertility/genetics , Male , Meiosis/genetics , Mice, Knockout , Mice, Transgenic , Pachytene Stage/genetics , Sertoli Cells/cytology , Spermatocytes/cytology , Testis/cytology
13.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135305

ABSTRACT

Ion channel-controlled cell volume regulation is of fundamental significance to the physiological function of sperm. In addition to volume regulation, LRRC8A-dependent volume-regulated anion channel (VRAC) activity is involved in cell cycle progression, insulin signaling, and cisplatin resistance. Nevertheless, the contribution of LRRC8A and its dependent VRAC activity in the germ cell lineage remain unknown. By utilizing a spontaneous Lrrc8a mouse mutation (c.1325delTG, p.F443*) and genetically engineered mouse models, we demonstrate that LRRC8A-dependent VRAC activity is essential for male germ cell development and fertility. Lrrc8a-null male germ cells undergo progressive degeneration independent of the apoptotic pathway during postnatal testicular development. Lrrc8a-deficient mouse sperm exhibit multiple morphological abnormalities of the flagella (MMAF), a feature commonly observed in the sperm of infertile human patients. Importantly, we identified a human patient with a rare LRRC8A hypomorphic mutation (c.1634G>A, p.Arg545His) possibly linked to Sertoli cell-only syndrome (SCOS), a male sterility disorder characterized by the loss of germ cells. Thus, LRRC8A is a critical factor required for germ cell development and volume regulation in the mouse, and it might serve as a novel diagnostic and therapeutic target for SCOS patients.


Subject(s)
Flagella/pathology , Infertility, Male/genetics , Membrane Proteins/genetics , Adult , Animals , Anions/metabolism , Biological Transport, Active/genetics , Biomarkers/analysis , Case-Control Studies , Cell Survival/genetics , China , Disease Models, Animal , Female , Healthy Volunteers , Humans , Infertility, Male/diagnosis , Infertility, Male/pathology , Ion Transport/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mutation , Sperm Motility/genetics , Spermatozoa/cytology , Spermatozoa/pathology , Testis/pathology
14.
Reprod Toxicol ; 69: 150-158, 2017 04.
Article in English | MEDLINE | ID: mdl-28238932

ABSTRACT

Exposure of rodents to the Sertoli cell (SC) toxicant mono-(2-ethylhexyl) phthalate (MEHP) has been reported to trigger an infiltration of macrophages into the testis in an age- and species-dependent manner. Here we challenge the hypothesis that the peripubertal rat-specific infiltration of macrophages after MEHP exposure is due, in part, to an increase in SC-specific inflammatory cytokine expression. To rule out that germ cell(GC) apoptosis itself is responsible for macrophage recruitment, rats were exposed to a direct GC toxicant, methoxyacetic acid (MAA), but no infiltration of macrophages was observed. Next, mRNA levels of inflammatory cytokines were evaluated after MEHP exposure. IL-1α, IL-6, and MCP-1 expression were increased in vivo and correlated with macrophage infiltration in a species-specific manner. Additionally, IL-6 and MCP-1 expression was increased in SC-GC co-cultures and ASC-17D SCs. These results indicate that MEHP-injury in pubertal rats specifically stimulates secretion of pro-inflammatory cytokines and alters the immune microenvironment.


Subject(s)
Cytokines/genetics , Diethylhexyl Phthalate/analogs & derivatives , Endocrine Disruptors/toxicity , Sertoli Cells/drug effects , Acetates/toxicity , Animals , Apoptosis/drug effects , Cell Line , Cells, Cultured , Diethylhexyl Phthalate/toxicity , Macrophages/drug effects , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism , Rats, Inbred F344 , Sertoli Cells/immunology , Species Specificity
15.
Exp Biol Med (Maywood) ; 241(4): 367-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26515141

ABSTRACT

Major progress in deciphering the role of the E3 ligase, ITCH, in animal physiology has come from the generation and identification of Itch loss-of-function mutant mice (itchy). Mutant mice display an autoimmune-like phenotype characterized by chronic dermatitis, which has been attributed to increased levels of ITCH target proteins (e.g. transcription factors JUNB and CJUN) in T cells. Autoimmune disorders also exist in humans with Itch frameshift mutations resulting in loss of functional ITCH protein. Recent phenotypic analysis of male itchy mice revealed reduced sperm production, although cross breeding experiments showed no difference in litter size when male itchy mice were bred to wild type females. However, a reduction in litter sizes did occur when itchy females were bred to wild type males. Based on these results, characterization of female reproductive function in itchy mice was performed. Developmental analysis of fetuses at gestational day 18.5, cytological evaluation of estrous cyclicity, histopathological analysis of ovaries, and protein analysis were used to investigate the itchy reproductive phenotype. Gross skeletal and soft tissue analysis of gestational day 18.5 itchy fetuses indicated no gross developmental deformities. Itchy females had reduced implantation sites, decreased corpora lutea, and increased estrous cycle length due to increased number of days in estrus compared to controls. Alterations in the expression of prototypical ITCH targets in the ovaries were not indicated, suggesting that an alteration in an as yet defined ovary-specific ITCH substrate or interaction with the altered immune system likely accounts for the disruption of female reproduction. This report indicates the importance of the E3 ligase, ITCH, in female reproduction.


Subject(s)
Estrous Cycle , Reproduction , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Humans , Male , Mice, Inbred C57BL
16.
J Androl ; 26(1): 136-45, 2005.
Article in English | MEDLINE | ID: mdl-15611578

ABSTRACT

Exposure to cisplatin results in impaired spermatogenesis, azoospermia, and, sometimes, permanent infertility in male patients. The mechanism(s) by which cisplatin induces damage to testicular cells is poorly understood. We previously reported that acute exposure to cisplatin results in elevated germ cell apoptotic rates and that this indicates long-term damage to the seminiferous epithelium. Here, we present data that implicate an injury to Sertoli cells as a possible mechanism to explain an elevated rate of germ cell apoptosis and consequent infertility. Normal adult C57/Bl/6J mice were exposed to 1, 2, or 4 rounds of 1, 2.5, or 5 mg/kg cisplatin in a regimen designed to resemble clinical chemotherapeutic exposure (1 injection daily for 5 days with a recovery phase of 16 days between cycles). A dose-dependent reduction in testicular weight due to germ cell loss was observed. While exposure to 1 mg/kg caused only temporary germ cell depletion, higher doses (2.5 and 5 mg/kg) revealed widespread testicular atrophy as evidenced by gaps in the epithelium due to cytoplasmic vacuolization and loss of differentiating germ cells. Although the acute loss of germ cells by apoptosis can result in temporary infertility, the testis has the ability to repopulate itself with mature cells, provided the stem germ cell population remains unharmed. Here, we demonstrate that a sustained disruption of spermatogenesis occurs despite the continued presence of stem spermatogonia in the seminiferous epithelium. These results suggest that cisplatin-induced germ cell loss may occur, in part, as a result of Sertoli cell injury-dependent alterations in germ cell microenvironment.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Spermatogenesis/drug effects , Testis/drug effects , Animals , Dose-Response Relationship, Drug , Immunohistochemistry , Male , Mice , Time Factors
17.
Toxicol Lett ; 139(1): 1-10, 2003 Mar 20.
Article in English | MEDLINE | ID: mdl-12595153

ABSTRACT

We have previously reported that the Fas/Apo-1/CD95-mediated apoptosis-inducing signaling system participates in the initiation of toxicant-induced testicular germ cell apoptosis. The contribution of Fas-mediated signaling is especially evident in the initiation of germ cell apoptosis after mono-(2-ethylhexyl)phthalate (MEHP)-induced Sertoli cell injury. In previous work, we demonstrated that the incidence of germ cell apoptosis after MEHP exposure is significantly reduced in B6.SMNC3H-Fas(gld,gld) (gld) mice that express a dysfunctional form of the FasL protein (the associated ligand that activates Fas). This has led to the hypothesis that activation of the Fas-mediated signaling pathway is a common mechanism for the initiation of germ cell apoptosis after toxicant-induced Sertoli cell injury. To test this hypothesis, we evaluated the sensitivity of testicular germ cells of wild-type, gld- and Fas-deficient CBA/KlJms-Tnfrsf6lpr-cg((lpr-cg)) (lpr(cg)) mice to undergo apoptosis after exposure to the Sertoli cell toxicant nitrobenzene (NB). Adult, 8-week-old gld mice treated with a single oral dose of NB (800 mg/kg) were observed to have a higher apoptotic index (AI; 66.1+/-1.3) 24 h after exposure as compared with the wild-type C57BL/6 (C57) mice (50.4+/-1.8). Similarly, 8-week-old lpr(cg) mice treated with NB displayed a higher AI 24 h after exposure (45.1+/-4.6) as compared with the wild-type CBA/KlJms (CBA) mice (32.1+/-3.8). Interestingly, exposure of both peri-pubertal 4-week-old C57 and gld mice showed a similar increase in the incidence of germ cell apoptosis after NB (600 mg/kg) exposure. Taken together, these findings indicate that Fas-mediated signaling is not required for NB-induced germ cell apoptosis and imply that a dysfunctional Fas signaling system sensitizes adult mice to NB-induced germ cell elimination.


Subject(s)
Apoptosis/drug effects , Diethylhexyl Phthalate/analogs & derivatives , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Nitrobenzenes/pharmacology , Spermatozoa/drug effects , Testis/drug effects , fas Receptor/metabolism , Aging , Animals , Diethylhexyl Phthalate/pharmacology , Fas Ligand Protein , Gene Deletion , Gene Expression Regulation , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nitrobenzenes/toxicity , Sertoli Cells/drug effects , Sertoli Cells/pathology , Signal Transduction , Spermatozoa/pathology , Testis/pathology , fas Receptor/genetics
18.
Toxicol Lett ; 135(3): 167-83, 2002 Oct 05.
Article in English | MEDLINE | ID: mdl-12270675

ABSTRACT

A symposium was held at the 41st annual meeting of the Society of Toxicology with presentations that emphasized novel molecular and cellular pathways that modulate the response to testicular toxicants. The first two presentations described cellular alterations after exposure to the Sertoli cell toxicant, mono-(2-ethylhexyl) phthalate (MEHP). The expression of flamingo1, a G protei coupled receptor family member that may couple cell-cell adhesion to G protein-dependent signaling in Sertoli cells, was rapidly altered after MEHP exposure. Sertoli cell associated flamingo1 immunostaining was redistributed early (within 2 h) after MEHP exposure and disappeared by 12 h, suggesting that flamingo1 is a proximal phthalate target. MEHP was also described to alter the expression and activity of the newly identified death receptors DR4, 5 and 6 in the testis. The differential cellular changes in the levels of DR4, 5 and 6 after phthalate exposure suggested that they may act as surrogates or in concert with the widely described Fas-signaling pathway in the initiation of germ cell apoptosis after MEHP exposure. The next two presentations focused on revealing mechanisms that may explain the persistent post-exposure testicular atrophy that is observed in rodents after a variety of chemical or physical insults (radiation, chemotherapeutics, toxicants) and possible strategies to re-initiate spermatogenesis in the atrophic testis. Hormonal manipulations that lower testosterone and serum FSH levels allow for re-initiation of spermatogonial development. Recent investigation of additional models of persistent atrophy such as mutant mice, the aged Brown Norway rat, EDS-induced Leydig cell deficient rat, and primates, have broadened insight into the mechanisms responsible for persistent atrophy. The last presentation described the use of cDNA arrays in the investigation of cellular elements and mechanisms responsible for disruption of spermatogenesis by the drinking water disinfectant bromochloroacetic acid (BCA). A custom mouse testis cDNA array interrogating 950 genes was used for analysis of testis mRNA. BCA altered the expression of 53 of the 950 genes, including two encoding sperm proteins known to be significant for male fertility, and other genes involved in spermatogenesis, stress response, and cell communication/adhesion. These observations strengthen the hypothesis that BCA disrupts spermatogenesis by altering the process of spermiogenesis.


Subject(s)
Hazardous Substances/pharmacology , Hazardous Substances/toxicity , Testis/drug effects , Animals , Apoptosis/drug effects , Cadherins/metabolism , Male , Signal Transduction/drug effects , Spermatozoa/drug effects , Testis/metabolism , Testis/pathology , fas Receptor/metabolism
19.
Spermatogenesis ; 4(2): e979110, 2014.
Article in English | MEDLINE | ID: mdl-26413394

ABSTRACT

After exposure to toxicants, degenerating germ cells represents the most common testicular histopathological alteration, regardless of the mechanism of toxicity. Therefore, deciphering the primary toxicant cellular target and mechanism of action can be extremely difficult. However, most testicular toxicants display a cell-specific and a stage-specific pattern of damage, which is the best evidence for identifying the primary cellular target (i.e. germ cell, Sertoli cell, peritubular myoid cell, or Leydig cell). Some toxicant-induced Sertoli cell injury presents with germ cell apoptosis occurring primarily in spermatocytes in rats in stages XI-XIV, I and II. Although some toxicants result in spermatid degeneration and apoptosis, it is still unclear if spermatid apoptosis is a result of Sertoli cell-selective apoptosis or a direct effect of toxicants on spermatids, therefore if this is seen as the earliest change, one cannot infer the mechanism of apoptosis. This review summarizes some of the distinguishing features of Sertoli cell-induced germ cell apoptosis and the associated mechanisms of cell death to provide the toxicologist observing similar cell death, with evidence about a potential mode of action.

20.
Toxicol Lett ; 227(2): 99-112, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24704392

ABSTRACT

A typical clinical cis-diamminedichloroplatinum(II) (cisplatin) dosing regimen consists of repeated treatment cycles followed by a recovery period. While effective, this dosing structure results in a prolonged, often permanent, infertility in men. Spermatogonial stem cells (SSCs) are theoretically capable of repopulating the seminiferous tubules after exposure has ceased. We propose that an altered spermatogonial environment during recovery from the initial treatment cycle drives an increase in SSC mitotic cell activity, rendering the SSC pool increasingly susceptible to cisplatin-induced injury from subsequent cycles. To test this hypothesis, the undifferentiated spermatogonia population and niche of the adult mouse (C57/BL/6J) were examined during the recovery periods of a clinically-relevant cisplatin exposure paradigm. Histological examination revealed a disorganization of spermatogenesis correlating with the number of exposure cycles. Quantification of terminal deoxynucleotidyl transferase-mediated digoxigenin-dUTP nick end labeling (TUNEL) staining indicated an increase in apoptotic frequency following exposure. Immunohistochemical examination of Foxo1 and incorporated BrdU showed an increase in the undifferentiated spermatogonial population and mitotic activity in the recovery period in mice exposed to one cycle, but not two cycles of cisplatin. Immunohistochemical investigation of glial cell line-derived neurotrophic factor (GDNF) revealed an increase in production along the basal Sertoli cell membrane throughout the recovery period in all treatment groups. Taken together, these data establish that the impact of cisplatin exposure on the functional stem cell pool and niche correlates with: (1) the number of dosing cycles; (2) mitotic activity of early germ cells; and (3) alterations in the basal Sertoli cell GDNF expression levels after cisplatin-induced testicular injury.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cisplatin/administration & dosage , Cisplatin/toxicity , Spermatogonia/cytology , Spermatogonia/drug effects , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Testis/cytology , Testis/drug effects , Testis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL